1
|
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
|
2
|
Stephenson KA, Peters P, Rae MG, O'Malley D. Astrocyte proliferation in the hippocampal dentate gyrus is suppressed across the lifespan of dystrophin-deficient mdx mice. Exp Physiol 2025; 110:585-598. [PMID: 39792584 PMCID: PMC11963898 DOI: 10.1113/ep092150] [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: 07/11/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
Absence of the structural protein, dystrophin, results in the neuromuscular disorder Duchenne Muscular Dystrophy (DMD). In addition to progressive skeletal muscle dysfunction, this multisystemic disorder can also result in cognitive deficits and behavioural changes that are likely to be consequences of dystrophin loss from central neurons and astrocytes. Dystrophin-deficient mdx mice exhibit decreases in grey matter volume in the hippocampus, the brain region that encodes and consolidates memories, and this is exacerbated with ageing. To understand changes in cellular composition that might underpin these age-related developments, we have compared neurogenesis and the prevalence of immunofluorescently identified newly born and mature neurons, astrocytes and microglia in the dentate gyrus of mdx and wild-type mice at 2, 4, 8 and 16 months of age. The number of adult-born neurons was suppressed in the dentate gyrus subgranular zone of 2-month-old mdx mice. However, the numbers of granule cells and GABAA receptor, alpha 1-expressing cells were similar in wild-type and mdx mice at all ages. Strikingly, the numbers of astrocytes, particularly in the dentate gyrus molecular layer, were suppressed in mdx mice at all time points. Thus, dystrophin loss was associated with reduced hippocampal neurogenesis in early life but did not impact the prevalence of mature neurons across the lifespan of mdx mice. In contrast, normal age-related dentate gyrus astrocyte proliferation was suppressed in dystrophic mice. Astrocytes are the most abundant cell type in the brain and are crucial in supporting neuronal function, such that loss of these cells is likely to contribute to hippocampal dysfunction reported in mdx mice.
Collapse
Affiliation(s)
| | - Polly Peters
- Department of Physiology, School of MedicineUniversity College CorkCorkIreland
| | - Mark G. Rae
- Department of Physiology, School of MedicineUniversity College CorkCorkIreland
| | - Dervla O'Malley
- Department of Physiology, School of MedicineUniversity College CorkCorkIreland
| |
Collapse
|
3
|
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
|
4
|
Komaki H, Takeshita E, Kunitake K, Ishizuka T, Shimizu-Motohashi Y, Ishiyama A, Sasaki M, Yonee C, Maruyama S, Hida E, Aoki Y. Phase 1/2 trial of brogidirsen: Dual-targeting antisense oligonucleotides for exon 44 skipping in Duchenne muscular dystrophy. Cell Rep Med 2025; 6:101901. [PMID: 39793573 PMCID: PMC11866436 DOI: 10.1016/j.xcrm.2024.101901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/01/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle disorder caused by mutations in the DMD gene, leading to dystrophin deficiency. Antisense oligonucleotide (ASO)-mediated exon skipping offers potential by partially restoring dystrophin, though current therapies remain mutation specific with limited efficacy. To overcome those limitations, we developed brogidirsen, a dual-targeting ASO composed of two directly connected 12-mer sequences targeting exon 44 using phosphorodiamidate morpholino oligomers. An open-label, dose-escalation, phase 1/2 trial assessed the safety, pharmacokinetics, and efficacy of brogidirsen in six ambulant patients with DMD amenable to exon 44 skipping. Following dose escalation, extended 24-week treatment with 40 mg/kg and 80 mg/kg yielded dose-dependent increases in dystrophin (16.63% and 24.47% of normal). Functional assessments indicated motor stabilization, and plasma proteomics revealed reductions in peptidyl arginine deiminase 2 (PADI2), titin (TTN), and myomesin 2 (MYOM2), highlighting potential biomarkers. Brogidirsen's efficacy was supported in vitro using urine-derived cells from patients with DMD. These promising results warrant a subsequent trial for DMD. This study was registered at ClinicalTrials.gov (NCT04129294).
Collapse
Affiliation(s)
- Hirofumi Komaki
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8551, Japan; Translational Medical Center, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8502, Japan
| | - Eri Takeshita
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8551, Japan
| | - Katsuhiko Kunitake
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8502, Japan
| | - Takami Ishizuka
- Translational Medical Center, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8502, Japan
| | - Yuko Shimizu-Motohashi
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8551, Japan
| | - Akihiko Ishiyama
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8551, Japan
| | - Masayuki Sasaki
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8551, Japan
| | - Chihiro Yonee
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City, Kagoshima 890-8520, Japan
| | - Shinsuke Maruyama
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City, Kagoshima 890-8520, Japan
| | - Eisuke Hida
- Department of Biostatistics and Data Science, Graduate School of Medicine, Osaka University, Suita-Shi, Osaka 565-0871, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8502, Japan.
| |
Collapse
|
5
|
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
|
6
|
Govaarts R, Doorenweerd N, Najac CF, Broek EM, Tamsma ME, Hollingsworth KG, Niks EH, Ronen I, Straub V, Kan HE. Probing diffusion of water and metabolites to assess white matter microstructure in Duchenne muscular dystrophy. NMR IN BIOMEDICINE 2024; 37:e5212. [PMID: 39005110 DOI: 10.1002/nbm.5212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 05/15/2024] [Accepted: 06/15/2024] [Indexed: 07/16/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive X-linked neuromuscular disorder caused by the absence of functional dystrophin protein. In addition to muscle, dystrophin is expressed in the brain in both neurons and glial cells. Previous studies have shown altered white matter microstructure in patients with DMD using diffusion tensor imaging (DTI). However, DTI measures the diffusion properties of water, a ubiquitous molecule, making it difficult to unravel the underlying pathology. Diffusion-weighted spectroscopy (DWS) is a complementary technique which measures diffusion properties of cell-specific intracellular metabolites. Here we performed both DWS and DTI measurements to disentangle intra- and extracellular contributions to white matter changes in patients with DMD. Scans were conducted in patients with DMD (15.5 ± 4.6 y/o) and age- and sex-matched healthy controls (16.3 ± 3.3 y/o). DWS measurements were obtained in a volume of interest (VOI) positioned in the left parietal white matter. Apparent diffusion coefficients (ADCs) were calculated for total N-acetylaspartate (tNAA), choline compounds (tCho), and total creatine (tCr). The tNAA/tCr and tCho/tCr ratios were calculated from the non-diffusion-weighted spectrum. Mean diffusivity (MD), radial diffusivity (RD), axial diffusivity (AD), and fractional anisotropy of water within the VOI were extracted from DTI measurements. DWS and DTI data from patients with DMD (respectively n = 20 and n = 18) and n = 10 healthy controls were included. No differences in metabolite ADC or in concentration ratios were found between patients with DMD and controls. In contrast, water diffusion (MD, t = -2.727, p = 0.011; RD, t = -2.720, p = 0.011; AD, t = -2.715, p = 0.012) within the VOI was significantly higher in patients compared with healthy controls. Taken together, our study illustrates the potential of combining DTI and DWS to gain a better understanding of microstructural changes and their association with disease mechanisms in a clinical setting.
Collapse
Affiliation(s)
- Rosanne Govaarts
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Duchenne Centre Netherlands, Leiden, The Netherlands
| | - Nathalie Doorenweerd
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Chloé F Najac
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emma M Broek
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maud E Tamsma
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kieren G Hollingsworth
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Duchenne Centre Netherlands, Leiden, The Netherlands
| | - Itamar Ronen
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, Brighton, UK
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Hermien E Kan
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Duchenne Centre Netherlands, Leiden, The Netherlands
| |
Collapse
|
7
|
Li Z, Abram L, Peall KJ. Deciphering the Pathophysiological Mechanisms Underpinning Myoclonus Dystonia Using Pluripotent Stem Cell-Derived Cellular Models. Cells 2024; 13:1520. [PMID: 39329704 PMCID: PMC11430605 DOI: 10.3390/cells13181520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 09/28/2024] Open
Abstract
Dystonia is a movement disorder with an estimated prevalence of 1.2% and is characterised by involuntary muscle contractions leading to abnormal postures and pain. Only symptomatic treatments are available with no disease-modifying or curative therapy, in large part due to the limited understanding of the underlying pathophysiology. However, the inherited monogenic forms of dystonia provide an opportunity for the development of disease models to examine these mechanisms. Myoclonus Dystonia, caused by SGCE mutations encoding the ε-sarcoglycan protein, represents one of now >50 monogenic forms. Previous research has implicated the involvement of the basal ganglia-cerebello-thalamo-cortical circuit in dystonia pathogenesis, but further work is needed to understand the specific molecular and cellular mechanisms. Pluripotent stem cell technology enables a patient-derived disease modelling platform harbouring disease-causing mutations. In this review, we discuss the current understanding of the aetiology of Myoclonus Dystonia, recent advances in producing distinct neuronal types from pluripotent stem cells, and their application in modelling Myoclonus Dystonia in vitro. Future research employing pluripotent stem cell-derived cellular models is crucial to elucidate how distinct neuronal types may contribute to dystonia and how disruption to neuronal function can give rise to dystonic disorders.
Collapse
Affiliation(s)
- Zongze Li
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; (Z.L.); (L.A.)
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Laura Abram
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; (Z.L.); (L.A.)
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Kathryn J. Peall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; (Z.L.); (L.A.)
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
8
|
Yslas AR, Park R, Nishimura N, Lee E. Monomeric and oligomeric amyloid-β cause distinct Alzheimer's disease pathophysiological characteristics in astrocytes in human glymphatics-on-chip models. LAB ON A CHIP 2024; 24:3826-3839. [PMID: 39037244 PMCID: PMC11302770 DOI: 10.1039/d4lc00287c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) is marked by the aggregation of extracellular amyloid-β (Aβ) and astrocyte dysfunction. For Aβ oligomers or aggregates to be formed, there must be Aβ monomers present; however, the roles of monomeric Aβ (mAβ) and oligomeric Aβ (oAβ) in astrocyte pathogenesis are poorly understood. We cultured astrocytes in a brain-mimicking three-dimensional (3D) extracellular matrix and revealed that both mAβ and oAβ caused astrocytic atrophy and hyper-reactivity, but showed distinct Ca2+ changes in astrocytes. This 3D culture evolved into a microfluidic glymphatics-on-chip model containing astrocytes and endothelial cells with the interstitial fluid (ISF). The glymphatics-on-chip model not only reproduced the astrocytic atrophy, hyper-reactivity, and Ca2+ changes induced by mAβ and oAβ, but recapitulated that the components of the dystrophin-associated complex (DAC) and aquaporin-4 (AQP4) were properly maintained by the ISF, and dysregulated by mAβ and oAβ. Collectively, mAβ and oAβ cause distinct AD pathophysiological characteristics in the astrocytes.
Collapse
Affiliation(s)
- Aria R Yslas
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| | - Rena Park
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| | - Nozomi Nishimura
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Barboni MTS, Joachimsthaler A, Roux MJ, Nagy ZZ, Ventura DF, Rendon A, Kremers J, Vaillend C. Retinal dystrophins and the retinopathy of Duchenne muscular dystrophy. Prog Retin Eye Res 2022:101137. [DOI: 10.1016/j.preteyeres.2022.101137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
|
12
|
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
|
13
|
Simon M, Wang MX, Ismail O, Braun M, Schindler AG, Reemmer J, Wang Z, Haveliwala MA, O’Boyle RP, Han WY, Roese N, Grafe M, Woltjer R, Boison D, Iliff JJ. Loss of perivascular aquaporin-4 localization impairs glymphatic exchange and promotes amyloid β plaque formation in mice. Alzheimers Res Ther 2022; 14:59. [PMID: 35473943 PMCID: PMC9040291 DOI: 10.1186/s13195-022-00999-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 04/04/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Slowed clearance of amyloid β (Aβ) is believed to underlie the development of Aβ plaques that characterize Alzheimer's disease (AD). Aβ is cleared in part by the glymphatic system, a brain-wide network of perivascular pathways that supports the exchange of cerebrospinal and brain interstitial fluid. Glymphatic clearance, or perivascular CSF-interstitial fluid exchange, is dependent on the astroglial water channel aquaporin-4 (AQP4) as deletion of Aqp4 in mice slows perivascular exchange, impairs Aβ clearance, and promotes Aβ plaque formation. METHODS To define the role of AQP4 in human AD, we evaluated AQP4 expression and localization in a human post mortem case series. We then used the α-syntrophin (Snta1) knockout mouse model which lacks perivascular AQP4 localization to evaluate the effect that loss of perivascular AQP4 localization has on glymphatic CSF tracer distribution. Lastly, we crossed this line into a mouse model of amyloidosis (Tg2576 mice) to evaluate the effect of AQP4 localization on amyloid β levels. RESULTS In the post mortem case series, we observed that the perivascular localization of AQP4 is reduced in frontal cortical gray matter of subjects with AD compared to cognitively intact subjects. This decline in perivascular AQP4 localization was associated with increasing Aβ and neurofibrillary pathological burden, and with cognitive decline prior to dementia onset. In rodent studies, Snta1 gene deletion slowed CSF tracer influx and interstitial tracer efflux from the mouse brain and increased amyloid β levels. CONCLUSIONS These findings suggest that the loss of perivascular AQP4 localization may contribute to the development of AD pathology in human populations.
Collapse
Affiliation(s)
- Matthew Simon
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR USA
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR USA
| | - Marie Xun Wang
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA USA
| | - Ozama Ismail
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR USA
- Center for Advanced Biomedical Imaging, University College London, London, UK
| | - Molly Braun
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA USA
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, 1660 S Columbian Wy., Seattle, WA 98108 USA
| | - Abigail G. Schindler
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA USA
- VISN 20 Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA USA
| | - Jesica Reemmer
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR USA
| | - Zhongya Wang
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR USA
| | - Mariya A. Haveliwala
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, 1660 S Columbian Wy., Seattle, WA 98108 USA
| | - Ryan P. O’Boyle
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, 1660 S Columbian Wy., Seattle, WA 98108 USA
| | - Warren Y. Han
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, 1660 S Columbian Wy., Seattle, WA 98108 USA
| | - Natalie Roese
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR USA
| | - Marjorie Grafe
- Department of Pathology, Oregon Health & Science University, Portland, OR USA
| | - Randall Woltjer
- Department of Pathology, Oregon Health & Science University, Portland, OR USA
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ USA
| | - Jeffrey J. Iliff
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA USA
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, 1660 S Columbian Wy., Seattle, WA 98108 USA
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA USA
| |
Collapse
|
14
|
Passos-Bueno MR, Costa CIS, Zatz M. Dystrophin genetic variants and autism. DISCOVER MENTAL HEALTH 2022; 2:4. [PMID: 37861890 PMCID: PMC10501027 DOI: 10.1007/s44192-022-00008-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/07/2022] [Indexed: 10/21/2023]
Abstract
Loss-of-function variants in the dystrophin gene, a well-known cause of muscular dystrophies, have emerged as a mutational risk mechanism for autism spectrum disorder (ASD), which in turn is a highly prevalent (~ 1%) genetically heterogeneous neurodevelopmental disorder. Although the association of intellectual disability with the dystrophinopathies Duchenne (DMD) and Becker muscular dystrophy (BMD) has been long established, their association with ASD is more recent, and the dystrophin genotype-ASD phenotype correlation is unclear. We therefore present a review of the literature focused on the ASD prevalence among dystrophinopathies, the relevance of the dystrophin isoforms, and most particularly the relevance of the genetic background to the etiology of ASD in these patients. Four families with ASD-DMD/BMD patients are also reported here for the first time. These include a single ASD individual, ASD-discordant and ASD-concordant monozygotic twins, and non-identical ASD triplets. Notably, two unrelated individuals, which were first ascertained because of the ASD phenotype at ages 15 and 5 years respectively, present rare dystrophin variants still poorly characterized, suggesting that some dystrophin variants may compromise the brain more prominently. Whole exome sequencing in these ASD-DMD/BMD individuals together with the literature suggest, although based on preliminary data, a complex and heterogeneous genetic architecture underlying ASD in dystrophinopathies, that include rare variants of large and medium effect. The need for the establishment of a consortia for genomic investigation of ASD-DMD/BMD patients, which may shed light on the genetic architecture of ASD, is discussed.
Collapse
Affiliation(s)
- 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, Brazil.
| | - 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, Brazil
| | - 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, Brazil
| |
Collapse
|
15
|
Wood H. Evidence for altered astrocyte function in Duchenne muscular dystrophy. Nat Rev Neurol 2021; 18:1. [PMID: 34880471 DOI: 10.1038/s41582-021-00601-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|