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
|
Bu C, Zha Xi Q, Deng Y. Clinical and genetic analysis of a female child with duplications at 7p22.3p22.1 and Xp22.31p21.1: A case report. Medicine (Baltimore) 2025; 104:e41452. [PMID: 39928819 PMCID: PMC11813060 DOI: 10.1097/md.0000000000041452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/12/2025] Open
Abstract
RATIONALE Intellectual disability (ID) is a neurodevelopmental disorder with diverse etiologies. Chromosomal duplications are recognized as a common cause of ID, with manifestations typically milder than those associated with deletions. Duplications involving the short arm of chromosome 7 or the X chromosome have been linked to ID. Limited data exist on duplications at 7p22.1p22.3 and Xp22.31p21.1, leaving their clinical significance largely unexplored. This case report aims to expand the phenotype and genetics spectrum of 7p22 duplication syndrome and X-linked ID. Special attention should be paid to closely monitoring the pubertal progression of such patients. PATIENT CONCERNS A 9-year-10-month-old female was admitted to our hospital due to distinctive dysmorphic features and ID. DIAGNOSES Upon examination, features of craniofacial dysmorphism were observed, including micrognathia, a prominent lower lip, a thin upper lip, a flat nasal bridge, and hypertelorism. The child's pubertal development is progressing extremely rapidly; at under 10 years old, the breasts have already advanced to Tanner Stage 4. Her height was within the median range, but her bone age was advanced (12.1 years). Her full-scale intelligence quotient, assessed using the Wechsler Intelligence Scale for Children: 4th edition, was 41. INTERVENTIONS Whole exome sequencing identified a de novo duplication spanning overlapping regions at Xp21-22 and 7p22. This duplication encompasses several genes implicated in ID, including Duchenne muscular dystrophy, Aristaless-related homeobox, interleukin-1 receptor accessory protein like 1, adaptor-related protein complex 1 subunit sigma 2, and cyclin-dependent kinase-like 5. OUTCOMES A novel duplication in the Xp and 7p of a Chinese female child diagnosed with ID and dysmorphic features has been studied by whole exome sequencing analysis. The novel duplications were a large duplication located in the 7p22.1 to p22.3 region, spanning 12.07 Mb, and a large duplication located in the Xp22.31 to p21.1 region, spanning 24.7 Mb. LESSONS Our study underscores the importance of comprehensive clinical and genetic evaluation in individuals with duplication on the X chromosome and the terminal region of chromosome 7's short arm. We highlight the need for monitoring these patients for growth and sexual development issues. Our findings also suggest that chromosomal duplication can lead to severe clinical manifestations, emphasizing the critical role of genetic assessment in managing such cases.
Collapse
Affiliation(s)
- Chenchen Bu
- Department of Pediatrics, Tibet Maternal and Child Health Hospital, Lhasa, China
| | - Quzhen Zha Xi
- Department of Pediatrics, Tibet Maternal and Child Health Hospital, Lhasa, China
| | - Ying Deng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Disease of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
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
|
Nakamura A, Matsumura T, Ogata K, Mori-Yoshimura M, Takeshita E, Kimura K, Arahata H, Takeshima Y, Takahashi T, Ishigaki K, Awano H, Sugie K, Fujii T, Oi H, Komaki H. Clinical Characteristics of Patients With Becker Muscular Dystrophy Having Pathogenic Microvariants or Duplications. Neurol Genet 2025; 11:e200215. [PMID: 39712290 PMCID: PMC11661972 DOI: 10.1212/nxg.0000000000200215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/01/2024] [Indexed: 12/24/2024]
Abstract
Background and Objectives Becker muscular dystrophy (BMD) is an allelic disorder of Duchenne muscular dystrophy (DMD) in which pathogenic variants in DMD cause progressive worsening of motor dysfunction, muscle weakness and atrophy, and death due to respiratory and cardiac failure. BMD often has in-frame deletions that preserve the amino acid reading frame, but there are some cases with microvariants or duplications. In recent years, the importance of therapeutic development and care for BMD has been emphasized. Therefore, the purpose of this study was to understand the clinical characteristics of BMD patients with microvariants or duplications and to determine the genotype-phenotype relationship. Methods The study focused on patients with pathogenic microvariants or duplications in DMD who were ambulatory after 16 years of age or had specific muscle biopsy results between June 13, 2017, and March 31, 2023. Informed consent was obtained from the patients or their surrogates. Data concerning DMD variants, muscle biopsy findings, skeletal muscle, respiratory and cardiac function, and CNS involvement were collected and analyzed statistically. Results Thirty-three patients with BMD had pathogenic microvariants (missense variants, nonsense variants, splice site variants, and other microvariants), and 16 patients had in-frame duplications in DMD. Many patients with microvariants had abnormal ECG findings. The effect of variant type on patient outcomes varied. Regardless of the type of microvariant, skeletal muscle and respiratory dysfunction was more severe in mutants of the cysteine-rich/C-terminal domain than in rod domain mutants. On the other hand, there was no significant difference in the complication rate of CNS disorders among the 3 domains of dystrophin. Discussion Microvariant forms, in particular, tend to vary in clinical severity according to the site of the dystrophin protein mutation rather than the type of pathogenic variant. The results of this study may be useful for genetic counseling, care, and treatment of patients with BMD.
Collapse
Affiliation(s)
- Akinori Nakamura
- Department of Clinical Research and Department of Neurology, NHO Matsumoto Medical Center
| | | | - Katsuhisa Ogata
- Department of Neurology, NHO Higashisaitama National Hospital, Hasuda
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira
| | - Eri Takeshita
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira
| | - Koichi Kimura
- Department of Laboratory Medicine/Cardiology, The Institute of Medical Science, The University of Tokyo
| | - Hajime Arahata
- Department of Neurology, Neuro-Muscular Center, NHO Omuta Hospital
| | | | | | - Keiko Ishigaki
- Department of Pediatrics, Tokyo Women's Medical University School of Medicine
| | - Hiroyuki Awano
- Organization for Research Initiative and Promotion, Tottori University, Yonago
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University, Kashihara
| | - Tatsuya Fujii
- Department of Pediatrics, Shiga Medical Center for Children, Moriyama; and
| | - Hideki Oi
- Department of Clinical Data Science, Clinical Research and Education Promotion Division, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Hirofumi Komaki
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira
| |
Collapse
|
5
|
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
|
6
|
Upadhyay V, Ray S, Panja S, Saviola AJ, Maluf NK, Mallela KMG. Biophysical characterization of the dystrophin C-terminal domain: Dystrophin interacts differentially with dystrobrevin isoforms. J Biol Chem 2024; 300:108002. [PMID: 39551137 PMCID: PMC11719305 DOI: 10.1016/j.jbc.2024.108002] [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: 05/12/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) gene encodes dystrophin, a large multidomain protein. Its nonfunctionality leads to dystrophinopathies like DMD and Becker muscular dystrophy, for which no cure is yet available. A few therapies targeted towards specific mutations can extend the lifespan of patients, although with limited efficacy and high costs, emphasizing the need for more general treatments. Dystrophin's complex structure with poorly understood domains and the presence of multiple isoforms with varied expression patterns in different tissues pose challenges in therapeutic development. The C-terminal (CT) domain of dystrophin is less understood in terms of its structure-function, although it has been shown to perform important functional roles by interacting with another cytosolic protein, dystrobrevin. Dystrophin and dystrobrevin stabilize the sarcolemma membrane by forming a multiprotein complex called dystrophin-associated glycoprotein complex that is destabilized in DMD. Dystrobrevin has two major isoforms, alpha and beta, with tissue-specific expression patterns. Here, we characterize the CT domain of dystrophin and its interactions with the two dystrobrevin isoforms. We show that the CT domain is nonglobular and shows reversible urea denaturation as well as thermal denaturation. It interacts with dystrobrevin isoforms differentially, with differences in binding affinity and the mode of interaction. We further show that the amino acid differences in the CT region of dystrobrevin isoforms contribute to these differences. These results have implications for the stability of dystrophin-associated glycoprotein complex in different tissues and explain the differing symptoms associated with DMD patients affecting organs beyond the skeletal muscles.
Collapse
Affiliation(s)
- Vaibhav Upadhyay
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shashikant Ray
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Sudipta Panja
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Krishna M G Mallela
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
7
|
Alonge P, Gadaleta G, Urbano G, Lupica A, Di Stefano V, Brighina F, Torrente A. The Role of Brain Plasticity in Neuromuscular Disorders: Current Knowledge and Future Prospects. Brain Sci 2024; 14:971. [PMID: 39451985 PMCID: PMC11506792 DOI: 10.3390/brainsci14100971] [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: 08/14/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Increasing evidence shows an involvement of brain plasticity mechanisms in both motor and central manifestations of neuromuscular disorders (NMDs). These mechanisms could be specifically addressed with neuromodulation or rehabilitation protocols. The aim of this scoping review is to summarise the evidence on plasticity mechanisms' involvement in NMDs to encourage future research. Methods: A scoping review was conducted searching the PubMed and Scopus electronic databases. We selected papers addressing brain plasticity and central nervous system (CNS) studies through non-invasive brain stimulation techniques in myopathies, muscular dystrophies, myositis and spinal muscular atrophy. Results: A total of 49 papers were selected for full-text examination. Regardless of the variety of pathogenetic and clinical characteristics of NMDs, studies show widespread changes in intracortical inhibition mechanisms, as well as disruptions in glutamatergic and GABAergic transmission, resulting in altered brain plasticity. Therapeutic interventions with neurostimulation techniques, despite being conducted only anecdotally or on small samples, show promising results; Conclusions: despite challenges posed by the rarity and heterogeneity of NMDs, recent evidence suggests that synaptic plasticity may play a role in the pathogenesis of various muscular diseases, affecting not only central symptoms but also strength and fatigue. Key questions remain unanswered about the role of plasticity and its potential as a therapeutic target. As disease-modifying therapies advance, understanding CNS involvement in NMDs could lead to more tailored treatments.
Collapse
Affiliation(s)
- Paolo Alonge
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (P.A.); (A.L.); (V.D.S.); (A.T.)
| | - Giulio Gadaleta
- Neuromuscular Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.G.); (G.U.)
| | - Guido Urbano
- Neuromuscular Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.G.); (G.U.)
| | - Antonino Lupica
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (P.A.); (A.L.); (V.D.S.); (A.T.)
- U.O.C. Neurologia, Azienda Ospedaliera Papardo, 98121 Messina, Italy
| | - Vincenzo Di Stefano
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (P.A.); (A.L.); (V.D.S.); (A.T.)
| | - Filippo Brighina
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (P.A.); (A.L.); (V.D.S.); (A.T.)
| | - Angelo Torrente
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (P.A.); (A.L.); (V.D.S.); (A.T.)
| |
Collapse
|
8
|
Miranda R, Ceschi L, Le Verger D, Nagapin F, Edeline JM, Chaussenot R, Vaillend C. Social and emotional alterations in mice lacking the short dystrophin-gene product, Dp71. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:21. [PMID: 39182120 PMCID: PMC11344925 DOI: 10.1186/s12993-024-00246-x] [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: 03/13/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The Duchenne and Becker muscular dystrophies (DMD, BMD) are neuromuscular disorders commonly associated with diverse cognitive and behavioral comorbidities. Genotype-phenotype studies suggest that severity and risk of central defects in DMD patients increase with cumulative loss of different dystrophins produced in CNS from independent promoters of the DMD gene. Mutations affecting all dystrophins are nevertheless rare and therefore the clinical evidence on the contribution of the shortest Dp71 isoform to cognitive and behavioral dysfunctions is limited. In this study, we evaluated social, emotional and locomotor functions, and fear-related learning in the Dp71-null mouse model specifically lacking this short dystrophin. RESULTS We demonstrate the presence of abnormal social behavior and ultrasonic vocalization in Dp71-null mice, accompanied by slight changes in exploratory activity and anxiety-related behaviors, in the absence of myopathy and alterations of learning and memory of aversive cue-outcome associations. CONCLUSIONS These results support the hypothesis that distal DMD gene mutations affecting Dp71 may contribute to the emergence of social and emotional problems that may relate to the autistic traits and executive dysfunctions reported in DMD. The present alterations in Dp71-null mice may possibly add to the subtle social behavior problems previously associated with the loss of the Dp427 dystrophin, in line with the current hypothesis that risk and severity of behavioral problems in patients increase with cumulative loss of several brain dystrophin isoforms.
Collapse
Affiliation(s)
- Rubén Miranda
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France.
- Department of Psychobiology and Methodology in Behavioral Sciences, Universidad Complutense de Madrid, Ciudad Universitaria, 28040, Madrid, Spain.
| | - Léa Ceschi
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Delphine Le Verger
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Flora Nagapin
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Jean-Marc Edeline
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Rémi Chaussenot
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France.
| |
Collapse
|
9
|
Rumney RMH, Pomeroy J, Górecki DC. Investigating the Involvement of C-X-C Motif Chemokine 5 and P2X7 Purinoceptor in Ectopic Calcification in Mouse Models of Duchenne Muscular Dystrophy. J Cell Biochem 2024; 125:e30617. [PMID: 38924558 DOI: 10.1002/jcb.30617] [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: 03/11/2024] [Revised: 05/14/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Ectopic calcification of myofibers is an early pathogenic feature in patients and animal models of Duchenne muscular dystrophy (DMD). In previous studies using the Dmdmdx-βgeo mouse model, we found that the dystrophin-null phenotype exacerbates this abnormality and that mineralised myofibers are surrounded by macrophages. Furthermore, the P2X7 purinoceptor, functioning in immune cells offers protection against dystrophic calcification. In the present study, by exploring transcriptomic data from Dmdmdx mice, we hypothesised these effects to be mediated by C-X-C motif chemokine 5 (CXCL5) downstream of P2X7 activation. We found that CXCL5 is upregulated in the quadriceps muscles of Dmdmdx-βgeo mice compared to wild-type controls. In contrast, at the cell level, dystrophic (SC5) skeletal muscle cells secreted less CXCL5 chemokine than wild-type (IMO) controls. Although release from IMO cells was increased by P2X7 activation, this could not explain the elevated CXCL5 levels observed in dystrophic muscle tissue. Instead, we found that CXCL5 is released by dystrophin-null macrophages in response to P2X7 activation, suggesting that macrophages are the source of CXCL5 in dystrophic muscles. The effects of CXCL5 upon mineralisation were investigated using the Alizarin Red assay to quantify calcium deposition in vitro. In basal (low phosphate) media, CXCL5 increased calcification in IMO but not SC5 myoblasts. However, in cultures treated in high phosphate media, to mimic dysregulated phosphate metabolism occurring in DMD, CXCL5 decreased calcification in both IMO and SC5 cells. These data indicate that CXCL5 is part of a homoeostatic mechanism regulating intracellular calcium, that CXCL5 can be released by macrophages in response to the extracellular ATP damage-associated signal, and that CXCL5 can be part of a damage response to protect against ectopic calcification. This mechanism is affected by DMD gene mutations.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Mice
- Chemokine CXCL5/metabolism
- Chemokine CXCL5/genetics
- Disease Models, Animal
- Receptors, Purinergic P2X7/metabolism
- Receptors, Purinergic P2X7/genetics
- Calcinosis/metabolism
- Calcinosis/pathology
- Calcinosis/genetics
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mice, Inbred mdx
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Knockout
Collapse
Affiliation(s)
- Robin M H Rumney
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Joanna Pomeroy
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Dariusz C Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
10
|
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
|
11
|
Tobochnik S, Dorotan MKC, Ghosh HS, Lapinskas E, Vogelzang J, Reardon DA, Ligon KL, Bi WL, Smirnakis SM, Lee JW. Glioma genetic profiles associated with electrophysiologic hyperexcitability. Neuro Oncol 2024; 26:323-334. [PMID: 37713468 PMCID: PMC10836775 DOI: 10.1093/neuonc/noad176] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Distinct genetic alterations determine glioma aggressiveness, however, the diversity of somatic mutations contributing to peritumoral hyperexcitability and seizures over the course of the disease is uncertain. This study aimed to identify tumor somatic mutation profiles associated with clinically significant hyperexcitability. METHODS A single center cohort of adults with WHO grades 1-4 glioma and targeted exome sequencing (n = 1716) was analyzed and cross-referenced with a validated EEG database to identify the subset of individuals who underwent continuous EEG monitoring (n = 206). Hyperexcitability was defined by the presence of lateralized periodic discharges and/or electrographic seizures. Cross-validated discriminant analysis models trained exclusively on recurrent somatic mutations were used to identify variants associated with hyperexcitability. RESULTS The distribution of WHO grades and tumor mutational burdens were similar between patients with and without hyperexcitability. Discriminant analysis models classified the presence or absence of EEG hyperexcitability with an overall accuracy of 70.9%, regardless of IDH1 R132H inclusion. Predictive variants included nonsense mutations in ATRX and TP53, indel mutations in RBBP8 and CREBBP, and nonsynonymous missense mutations with predicted damaging consequences in EGFR, KRAS, PIK3CA, TP53, and USP28. This profile improved estimates of hyperexcitability in a multivariate analysis controlling for age, sex, tumor location, integrated pathologic diagnosis, recurrence status, and preoperative epilepsy. Predicted somatic mutation variants were over-represented in patients with hyperexcitability compared to individuals without hyperexcitability and those who did not undergo continuous EEG. CONCLUSION These findings implicate diverse glioma somatic mutations in cancer genes associated with peritumoral hyperexcitability. Tumor genetic profiling may facilitate glioma-related epilepsy prognostication and management.
Collapse
Affiliation(s)
- Steven Tobochnik
- Department of Neurology, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - Hia S Ghosh
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Emily Lapinskas
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jayne Vogelzang
- Department of Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - David A Reardon
- Department of Medical Oncology, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Keith L Ligon
- Department of Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Stelios M Smirnakis
- Department of Neurology, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jong Woo Lee
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Nakamura A, Matsumura T, Ogata K, Mori‐Yoshimura M, Takeshita E, Kimura K, Kawashima T, Tomo Y, Arahata H, Miyazaki D, Takeshima Y, Takahashi T, Ishigaki K, Kuru S, Wakisaka A, Awano H, Funato M, Sato T, Saito Y, Takada H, Sugie K, Kobayashi M, Ozasa S, Fujii T, Maegaki Y, Oi H, Tachimori H, Komaki H. Natural history of Becker muscular dystrophy: a multicenter study of 225 patients. Ann Clin Transl Neurol 2023; 10:2360-2372. [PMID: 37882106 PMCID: PMC10723226 DOI: 10.1002/acn3.51925] [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: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
OBJECTIVE Becker muscular dystrophy (BMD) is a milder variant of Duchenne muscular dystrophy (DMD), a lethal X-linked muscular disorder. Here, we aim to investigat the clinical involvement of skeletal, respiratory, cardiac, and central nervous systems in patients with BMD, as well as genotype-phenotype relationships. METHODS This nationwide cohort study investigated the clinical manifestations and genotype-phenotype relationships in 225 patients with BMD having in-frame deletion from 22 medical centers. The primary outcome was to elucidate the association of genotype with skeletal muscle, respiratory, cardiac, and central nervous system disorders. Descriptive statistics were used to analyze the data. RESULTS The average age of the subjects was 31.5 (range, 1-81) years. Initial symptoms of BMD were muscular (60%), followed by asymptomatic hypercreatine kinasemia (32.4%) and central nervous system disorders (5.3%). Gait disturbance was observed in 53.8% of patients and the average age at wheelchair introduction was 36.5 years. The ventilator introduction rate was 6.7% at an average age of 36.6 years. More than 30% of patients had an abnormal electrocardiogram and approximately 15% had heart failure symptoms. Cardiac function on echocardiography varied significantly among the patients. The frequencies of seizures and intellectual/developmental disability were 8.0% and 16.9%, respectively. Exon 45-47deletion (del) was the most common (22.6%), followed by exon 45-48del (13.1%). Patients with exon 45-49del patients demonstrated severe skeletal muscle damage. Patients with exon 45-47del and exon 45-55del patients did not require ventilator use. INTERPRETATION The study provides important prognostic information for patients and clinicians to establish therapy plans and to implement preventative medicine.
Collapse
Affiliation(s)
- Akinori Nakamura
- Department of NeurologyNHO Matsumoto Medical CenterMatsumotoJapan
| | | | - Katsuhisa Ogata
- Department of NeurologyNHO Higashisaitama National HospitalHasudaJapan
| | - Madoka Mori‐Yoshimura
- Department of NeurologyNational Center Hospital, National Center of Neurology and PsychiatryKodairaJapan
| | - Eri Takeshita
- Department of Child NeurologyNational Center Hospital, National Center of Neurology and PsychiatryKodairaJapan
| | - Koichi Kimura
- Department of Laboratory Medicine/CardiologyThe Institute of Medical Science, The University of TokyoMinato‐kuJapan
| | - Takahiro Kawashima
- Department of Information Medicine, National Center of Neurology and PsychiatryNational Institute of NeuroscienceKodairaJapan
| | - Yui Tomo
- Department of Clinical Data Science, Clinical Research & Education Promotion Division, National Center of Neurology and PsychiatryKodairaJapan
| | - Hajime Arahata
- Department of Neurology, Neuro‐Muscular CenterNHO Omuta National HospitalOmutaJapan
| | - Daigo Miyazaki
- Department of Medicine (Neurology and Rheumatology)Shinshu University School of MedicineMatsumotoJapan
| | - Yasuhiro Takeshima
- Department of PediatricsHyogo Medical University School of MedicineNishinomiyaJapan
| | | | - Keiko Ishigaki
- Department of PediatricsTokyo Women's Medical University School of MedicineShinjuku‐kuJapan
| | - Satoshi Kuru
- Department of NeurologyNHO Suzuka National HospitalSuzukaJapan
| | - Akiko Wakisaka
- Department of PediatricsNHO Iou National HospitalKanazawaJapan
| | - Hiroyuki Awano
- Research Initiative Center, Organization for Research Initiative and PromotionTottori UniversityYonagoJapan
| | - Michinori Funato
- Department of Pediatric NeurologyNHO Nagara Medical CenterNagaraJapan
| | - Tatsuharu Sato
- Department of PediatricsNagasaki University HospitalNagasakiJapan
| | - Yoshiaki Saito
- Department of PediatricsNational Rehabilitation Center for Children with DisabilitiesItabashiJapan
| | - Hiroto Takada
- Department of NeurologyNHO Aomori National HospitalAomoriJapan
| | - Kazuma Sugie
- Department of NeurologyNara Medical University School of MedicineKashiharaJapan
| | - Michio Kobayashi
- Department of NeurologyNHO Akita National HospitalYurihonjoJapan
| | - Shiro Ozasa
- Department of PediatricsKumamoto University HospitalKumamotoJapan
| | - Tatsuya Fujii
- Department of PediatricsShiga Medical Center for ChildrenMoriyamaJapan
| | - Yoshihiro Maegaki
- Division of Child Neurology, Department of Brain and Neurosciences, Faculty of MedicineTottori UniversityYonagoJapan
| | - Hideki Oi
- Department of Clinical Data Science, Clinical Research & Education Promotion Division, National Center of Neurology and PsychiatryKodairaJapan
| | - Hisateru Tachimori
- Department of Information Medicine, National Center of Neurology and PsychiatryNational Institute of NeuroscienceKodairaJapan
- Endowed Course of Health System InnovationKeio University School of MedicineTokyoJapan
| | - Hirofumi Komaki
- Department of Child NeurologyNational Center Hospital, National Center of Neurology and PsychiatryKodairaJapan
| |
Collapse
|
13
|
Andrews JG, Galindo MK, Thomas S, Mathews KD, Whitehead N. DMD Gene and Dystrophinopathy Phenotypes Associated With Mutations: A Systematic Review for Clinicians. J Clin Neuromuscul Dis 2023; 24:171-187. [PMID: 37219861 DOI: 10.1097/cnd.0000000000000436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
ABSTRACT The diagnosis of Duchenne and Becker muscular dystrophy (DBMD) is made by genetic testing in approximately 95% of cases. Although specific mutations can be associated with skeletal muscle phenotype, pulmonary and cardiac comorbidities (leading causes of death in Duchenne) have not been associated with Duchenne muscular dystrophy mutation type or location and vary within families. Therefore, identifying predictors for phenotype severity beyond frameshift prediction is important clinically. We performed a systematic review assessing research related to genotype-phenotype correlations in DBMD. While there are severity differences across the spectrum and within mild and severe forms of DBMD, few protective or exacerbating mutations within the dystrophin gene were reported. Except for intellectual disability, clinical test results reporting genotypic information are insufficient for clinical prediction of severity and comorbidities and the predictive validity is too low to be useful when advising families. Including expanded information coupled with proposed severity predictions in clinical genetic reports for DBMD is critical for improving anticipatory guidance.
Collapse
Affiliation(s)
- Jennifer G Andrews
- Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ
| | | | | | - Katherine D Mathews
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA; and
| | | |
Collapse
|
14
|
Pascual-Morena C, Cavero-Redondo I, Martínez-Vizcaíno V, Sequí-Domínguez I, Fernández-Bravo-Rodrigo J, Jiménez-López E. Dystrophin Genotype and Risk of Neuropsychiatric Disorders in Dystrophinopathies: A Systematic Review and Meta-Analysis. J Neuromuscul Dis 2023; 10:159-172. [PMID: 36565132 PMCID: PMC10041431 DOI: 10.3233/jnd-221586] [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: 12/24/2022]
Abstract
BACKGROUND Dystrophinopathies are associated with neuropsychiatric disorders due to alterations in dystrophin/DMD expression. OBJECTIVE The objective was to estimate the association of developmental disorders, autism spectrum disorders (ASD), attention deficit hyperactivity disorder (ADHD), depression, anxiety disorders, and obsessive-compulsive disorder with the dystrophin/DMD genotype in population with dystrophinopathies. METHODS Systematic searches of Medline, Scopus, Web of Science, and Cochrane Library were performed from inception to September 2022. We included observational studies in the population with Becker or Duchenne muscular dystrophies (BMD, DMD) that estimated the prevalence of these disorders according to Dp140 and/or Dp71 genotype. Meta-analysis of the prevalence ratio (PR) of genotype comparisons was conducted for each disorder. RESULTS Ten studies were included in the systematic review. In BMD, Dp140+ vs. Dp140- and Dp71+ vs. Dp71- were associated with developmental disorders with a PR of 0.11 (0.04, 0.34) and 0.22 (0.07, 0.67), respectively. In DMD, Dp140+/Dp71+ vs. Dp140- /Dp71- had a PR of 0.40 (0.28, 0.57), and Dp71+ vs. Dp71- had a PR of 0.47 (0.36, 0.63) for ADHD. However, there was no association of genotype with ASD, only a trend was observed for Dp71+ vs. Dp71-, with a PR of 0.61 (0.35, 1.06). Moreover, the data showed no association of these isoforms with emotional-related disorders. CONCLUSIONS In BMD, Dp140 and Dp71 could be associated with developmental disorders, while ADHD might be associated with the Dp71 genotype in DMD. Further research is needed regarding Dp140 and Dp71, especially in DMD for ASD.
Collapse
Affiliation(s)
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad deCastilla- La Mancha, Cuenca, Spain.,Facultadde Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Vicente Martínez-Vizcaíno
- Health and Social Research Center, Universidad deCastilla- La Mancha, Cuenca, Spain.,Facultadde Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | | | - Jaime Fernández-Bravo-Rodrigo
- Health and Social Research Center, Universidad deCastilla- La Mancha, Cuenca, Spain.,Pharmacy Service. Hospital Virgen de laLuz, Cuenca, Spain
| | - Estela Jiménez-López
- Health and Social Research Center, Universidad deCastilla- La Mancha, Cuenca, Spain
| |
Collapse
|
15
|
Pascual-Morena C, Cavero-Redondo I, Reina-Gutiérrez S, Saz-Lara A, López-Gil JF, Martínez-Vizcaíno V. Prevalence of Neuropsychiatric Disorders in Duchenne and Becker Muscular Dystrophies: A Systematic Review and Meta-analysis. Arch Phys Med Rehabil 2022; 103:2444-2453. [PMID: 35839922 DOI: 10.1016/j.apmr.2022.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/07/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To estimate the prevalence of neuropsychiatric disorders, including autism spectrum disorders (ASDs), attention-deficit hyperactivity disorder (ADHD), depression, anxiety disorders, and obsessive-compulsive disorder (OCD), in populations with Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). DATA SOURCES MEDLINE (via PubMed), Scopus, Web of Science, and Cochrane Library from inception to November 2021. STUDY SELECTION Observational studies of individuals with DMD or BMD that estimated the prevalence of ASDs, ADHD, depression, anxiety disorders, and OCD in each population. DATA EXTRACTION A random-effects meta-analysis was performed on each outcome and each population (ie, DMD, BMD). DATA SYNTHESIS Twenty-three studies were included in the meta-analysis. In DMD, there was a prevalence of 7.0% of ASDs, 18.0% of ADHD, 11.0% of depression, 24.0% of anxiety disorders, and 12.0% of OCD. Furthermore, in BMD, there was a prevalence of 6.0% of ASDs, 28.0% of ADHD, 7.0% of depression, 25.0% of anxiety disorders, and 7.0% of OCD. CONCLUSIONS The prevalence of these neuropsychiatric disorders is higher among patients with DMD or BMD than among the general population, and the presence of these disorders may negatively influence optimal medical management.
Collapse
Affiliation(s)
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain; Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Sara Reina-Gutiérrez
- 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
|
16
|
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
|
17
|
Tong C, Avilés L, Rayor LS, Mikheyev AS, Linksvayer TA. Genomic signatures of recent convergent transitions to social life in spiders. Nat Commun 2022; 13:6967. [PMID: 36414623 PMCID: PMC9681848 DOI: 10.1038/s41467-022-34446-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 10/25/2022] [Indexed: 11/24/2022] Open
Abstract
The transition from solitary to social life is a major phenotypic innovation, but its genetic underpinnings are largely unknown. To identify genomic changes associated with this transition, we compare the genomes of 22 spider species representing eight recent and independent origins of sociality. Hundreds of genes tend to experience shifts in selection during the repeated transition to social life. These genes are associated with several key functions, such as neurogenesis, behavior, and metabolism, and include genes that previously have been implicated in animal social behavior and human behavioral disorders. In addition, social species have elevated genome-wide rates of molecular evolution associated with relaxed selection caused by reduced effective population size. Altogether, our study provides unprecedented insights into the genomic signatures of social evolution and the specific genetic changes that repeatedly underpin the evolution of sociality. Our study also highlights the heretofore unappreciated potential of transcriptomics using ethanol-preserved specimens for comparative genomics and phylotranscriptomics.
Collapse
Affiliation(s)
- Chao Tong
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, 79409, USA.
| | - Leticia Avilés
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Linda S Rayor
- Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
| | - Alexander S Mikheyev
- Evolutionary Genomics Group, Research School of Biology, Australian National University, Canberra, 0200, Australia
| | - Timothy A Linksvayer
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, 79409, USA.
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
Nuclear transport and subcellular localization of the dystrophin Dp71 and Dp40 isoforms in the PC12 cell line. Biochem Biophys Res Commun 2022; 630:125-132. [PMID: 36155058 DOI: 10.1016/j.bbrc.2022.09.035] [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: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/21/2022]
Abstract
The shortest dystrophins, Dp71 and Dp40, are transcribed from the DMD gene through an internal promoter located in intron 62. These proteins are the main product of the DMD gene in the nervous system and have been involved in various functions related to cellular differentiation and proliferation as well as other cellular processes. Dp71 mRNA undergoes alternative splicing that results in different Dp71 protein isoforms. The subcellular localization of some of these isoforms in the PC12 cell line has been previously reported, and a differential subcellular distribution was observed, which suggests a particular role for each isoform. With the aim of obtaining information on their function, this study identified factors involved in the nuclear transport of Dp71 and Dp40 isoforms in the PC12 cell line. Cell cultures were treated with specific nuclear import/export inhibitors to determine the Dp71 isoform transport routes. The results showed that all isoforms of Dp71 and Dp40 included in the analysis have the ability to enter the cell nucleus through α/β importin, and the main route of nuclear export for Dp71 isoforms is through the exportin CRM1, which is not the case for Dp40.
Collapse
|
22
|
Ferrero A, Rossi M. Cognitive profile and neuropsychiatric disorders in Becker muscular dystrophy: A systematic review of literature. Neurosci Biobehav Rev 2022; 137:104648. [PMID: 35367224 DOI: 10.1016/j.neubiorev.2022.104648] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/15/2022] [Accepted: 03/29/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Brain co-morbidities in DMD are well-documented, less is known about the cognitive, behavioral and psychosocial functioning of patients with BMD. METHODS The systematic review was carried out on two databases (Pubmed and Scopus) according to the PRISMA guidelines. We included all research articles specific to BMD written after 1995. RESULTS Studies examining neuropsychological and neurobehavioral functioning in BMD are few and have several methods limitations. BMD population is characterized by high rates of cognitive impairment, with specific involvement of different cognitive areas. Unlike DMD, verbal skills are better preserved. Neurodevelopmental and emotional/behavioral disorders have great importance in BMD, due to their high prevalence. Lack of Dp140 or Dp71 can cause intellectual disability, these isoforms are probably responsible for the other brain-related comorbidities as well. DISCUSSION The results suggest that cognitive and neuropsychiatric comorbid symptoms may affect a significant proportion of BMD patients therefore it is important to mental health and neuropsychological screening. Finding tools for an adequate assessment is a priority in order to include brain outcome measures in clinical trials.
Collapse
Affiliation(s)
- Amanda Ferrero
- Department of Brain and Behavioural Sciences, Unit of Child Neurology and Psychiatry, University of Pavia, Pavia, Italy.
| | - Marta Rossi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
23
|
Stefano MED, Ferretti V, Mozzetta C. Synaptic alterations as a neurodevelopmental trait of Duchenne muscular dystrophy. Neurobiol Dis 2022; 168:105718. [PMID: 35390481 DOI: 10.1016/j.nbd.2022.105718] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/14/2023] Open
Abstract
Dystrophinopaties, e.g., Duchenne muscular dystrophy (DMD), Becker muscular dystrophy and X-linked dilated cardiomyopathy are inherited neuromuscular diseases, characterized by progressive muscular degeneration, which however associate with a significant impact on general system physiology. The more severe is the pathology and its diversified manifestations, the heavier are its effects on organs, systems, and tissues other than muscles (skeletal, cardiac and smooth muscles). All dystrophinopaties are characterized by mutations in a single gene located on the X chromosome encoding dystrophin (Dp427) and its shorter isoforms, but DMD is the most devasting: muscular degenerations manifests within the first 4 years of life, progressively affecting motility and other muscular functions, and leads to a fatal outcome between the 20s and 40s. To date, after years of studies on both DMD patients and animal models of the disease, it has been clearly demonstrated that a significant percentage of DMD patients are also afflicted by cognitive, neurological, and autonomic disorders, of varying degree of severity. The anatomical correlates underlying neural functional damages are established during embryonic development and the early stages of postnatal life, when brain circuits, sensory and motor connections are still maturing. The impact of the absence of Dp427 on the development, differentiation, and consolidation of specific cerebral circuits (hippocampus, cerebellum, prefrontal cortex, amygdala) is significant, and amplified by the frequent lack of one or more of its lower molecular mass isoforms. The most relevant aspect, which characterizes DMD-associated neurological disorders, is based on morpho-functional alterations of selective synaptic connections within the affected brain areas. This pathological feature correlates neurological conditions of DMD to other severe neurological disorders, such as schizophrenia, epilepsy and autistic spectrum disorders, among others. This review discusses the organization and the role of the dystrophin-dystroglycan complex in muscles and neurons, focusing on the neurological aspect of DMD and on the most relevant morphological and functional synaptic alterations, in both central and autonomic nervous systems, described in the pathology and its animal models.
Collapse
Affiliation(s)
- Maria Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy.
| | - Valentina Ferretti
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy c/o Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
24
|
Crawford AH, Hildyard JCW, Rushing SAM, Wells DJ, Diez-Leon M, Piercy RJ. Validation of DE50-MD dogs as a model for the brain phenotype of Duchenne muscular dystrophy. Dis Model Mech 2022; 15:dmm049291. [PMID: 35019137 PMCID: PMC8906169 DOI: 10.1242/dmm.049291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/21/2021] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), a fatal musculoskeletal disease, is associated with neurodevelopmental disorders and cognitive impairment caused by brain dystrophin deficiency. Dog models of DMD represent key translational tools to study dystrophin biology and to develop novel therapeutics. However, characterisation of dystrophin expression and function in the canine brain is lacking. We studied the DE50-MD canine model of DMD that has a missense mutation in the donor splice site of exon 50. Using a battery of cognitive tests, we detected a neurocognitive phenotype in DE50-MD dogs, including reduced attention, problem solving and exploration of novel objects. Through a combination of capillary immunoelectrophoresis, immunolabelling, quantitative PCR and RNAScope in situ hybridisation, we show that regional dystrophin expression in the adult canine brain reflects that of humans, and that the DE50-MD dog lacks full-length dystrophin (Dp427) protein expression but retains expression of the two shorter brain-expressed isoforms, Dp140 and Dp71. Thus, the DE50-MD dog is a translationally relevant pre-clinical model to study the consequences of Dp427 deficiency in the brain and to develop therapeutic strategies for the neurological sequelae of DMD.
Collapse
Affiliation(s)
- Abbe H. Crawford
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| | - Sophie A. M. Rushing
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Maria Diez-Leon
- Pathobiology and Population Sciences, Royal Veterinary College, London AL9 7TA, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| |
Collapse
|
25
|
Epileptic disorders in Becker and Duchenne muscular dystrophies: a systematic review and meta-analysis. J Neurol 2022; 269:3461-3469. [PMID: 35229191 DOI: 10.1007/s00415-022-11040-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 10/19/2022]
Abstract
Dystrophin alterations in the brain have been associated with an increased risk of epilepsy in Becker and Duchenne muscular dystrophies (BMD and DMD). Moreover, an association between the mutation site and the risk of epilepsy is not ruled out. The aim of this systematic review and meta-analysis was to estimate the prevalence of epilepsy in BMD and DMD populations and to establish a possible association between the site of mutation in the dystrophin gene and the risk of epilepsy. Systematic searches of Medline, Scopus, Web of Science, and Cochrane Library were conducted to identify relevant studies published from inception to January 2022. Observational studies of participants with BMD/DMD estimating the prevalence of epilepsy were included. The main outcome was the prevalence of epilepsy, and the secondary outcome was the prevalence ratio considering genotype. A random effects meta-analysis was performed for the prevalence of epilepsy. Eight studies were included in the systematic review and meta-analysis. The prevalence of epilepsy was 7% (95% CI 3-11%) in BMD, 5% (95% CI 2-8%) in DMD, and 5% (95% CI 3-7%) in the overall estimate. No association was observed between mutation site and the prevalence of epilepsy. BMD/DMD is strongly associated with the prevalence of epilepsy, with a higher prevalence in BMD/DMD populations than in the general population, probably owing to alterations in Dp427. The current evidence does not support the hypothesis that Dp140 or Dp71 affect epilepsy risk.
Collapse
|
26
|
Fujimoto T, Yaoi T, Nakano K, Arai T, Okamura T, Itoh K. Generation of dystrophin short product-specific tag-insertion mouse: distinct Dp71 glycoprotein complexes at inhibitory postsynapse and glia limitans. Cell Mol Life Sci 2022; 79:109. [PMID: 35098363 PMCID: PMC11071725 DOI: 10.1007/s00018-022-04151-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/24/2021] [Accepted: 01/14/2022] [Indexed: 11/03/2022]
Abstract
Duchenne muscular dystrophy (DMD), the most severe form of dystrophinopathies, is a fatal X-linked recessive neuromuscular disorder characterized by progressive muscle degeneration and various extents of intellectual disabilities. Physiological and pathological roles of the responsible gene, dystrophin, in the brain remain elusive due to the presence of multiple dystrophin products, mainly full-length dystrophin, Dp427, and the short product, Dp71. In this study, we generated a Dp71-specific hemagglutinin (HA) peptide tag-insertion mice to enable specific detection of intrinsic Dp71 expression by anti-HA-tag antibodies. Immunohistochemical detections in the transgenic mice demonstrated Dp71 expression not only at the blood-brain barrier, where astrocytic endfeet surround the microvessels, but also at the inhibitory postsynapse of hippocampal dentate granule neurons. Interestingly, hippocampal cornu ammonis (CA)1 pyramidal neurons were negative for Dp71, although Dp427 detected by anti-dystrophin antibody was clearly present at the inhibitory postsynapse, suggesting cell-type dependent dystrophin expressions. Precise examination using the primary hippocampal culture validated exclusive localization of Dp71 at the inhibitory postsynaptic compartment but not at the excitatory synapse in neurons. We further performed interactome analysis and found that Dp71 formed distinct molecular complexes, i.e. synapse-associated Dp71 interacted with dystroglycan (Dg) and dystrobrevinβ (Dtnb), whereas glia-associated Dp71 did with Dg and dystrobrevinα (Dtna). Thus, our data indicate that Dp71 and its binding partners are relevant to the inhibitory postsynaptic function of hippocampal granule neurons and the novel Dp71-transgenic mouse provides a valuable tool to understand precise physiological expressions and functions of Dp71 and its interaction proteins in vivo and in vitro.
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
| | - 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
| | - Tetsuya Arai
- 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
|
27
|
Overexpression of the dystrophins Dp40 and Dp40 L170P modifies neurite outgrowth and the protein expression profile of PC12 cells. Sci Rep 2022; 12:1410. [PMID: 35082358 PMCID: PMC8791958 DOI: 10.1038/s41598-022-05271-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/04/2022] [Indexed: 11/08/2022] Open
Abstract
Dp40 is ubiquitously expressed including the central nervous system. In addition to being present in the nucleus, membrane, and cytoplasm, Dp40 is detected in neurites and postsynaptic spines in hippocampal neurons. Although Dp40 is expressed from the same promoter as Dp71, its role in the cognitive impairment present in Duchenne muscular dystrophy patients is still unknown. Here, we studied the effects of overexpression of Dp40 and Dp40L170P during the neuronal differentiation of PC12 Tet-On cells. We found that Dp40 overexpression increased the percentage of PC12 cells with neurites and neurite length, while Dp40L170P overexpression decreased them compared to Dp40 overexpression. Two-dimensional gel electrophoresis analysis showed that the protein expression profile was modified in nerve growth factor-differentiated PC12-Dp40L170P cells compared to that of the control cells (PC12 Tet-On). The proteins α-internexin and S100a6, involved in cytoskeletal structure, were upregulated. The expression of vesicle-associated membrane proteins increased in differentiated PC12-Dp40 cells, in contrast to PC12-Dp40L170P cells, while neurofilament light-chain was decreased in both differentiated cells. These results suggest that Dp40 has an important role in the neuronal differentiation of PC12 cells through the regulation of proteins involved in neurofilaments and exocytosis of synaptic vesicles, functions that might be affected in PC12-Dp40L170P.
Collapse
|
28
|
Falzarano MS, Grilli A, Zia S, Fang M, Rossi R, Gualandi F, Rimessi P, El Dani R, Fabris M, Lu Z, Li W, Mongini T, Ricci F, Pegoraro E, Bello L, Barp A, Sansone VA, Hegde M, Roda B, Reschiglian P, Bicciato S, Selvatici R, Ferlini A. RNA-seq in DMD urinary stem cells recognized muscle-related transcription signatures and addressed the identification of atypical mutations by whole-genome sequencing. HGG ADVANCES 2022; 3:100054. [PMID: 35047845 PMCID: PMC8756543 DOI: 10.1016/j.xhgg.2021.100054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022] Open
Abstract
Urinary stem cells (USCs) are a non-invasive, simple, and affordable cell source to study human diseases. Here we show that USCs are a versatile tool for studying Duchenne muscular dystrophy (DMD), since they are able to address RNA signatures and atypical mutation identification. Gene expression profiling of DMD individuals' USCs revealed a profound deregulation of inflammation, muscle development, and metabolic pathways that mirrors the known transcriptional landscape of DMD muscle and worsens following USCs' myogenic transformation. This pathogenic transcription signature was reverted by an exon-skipping corrective approach, suggesting the utility of USCs in monitoring DMD antisense therapy. The full DMD transcript profile performed in USCs from three undiagnosed DMD individuals addressed three splicing abnormalities, which were decrypted and confirmed as pathogenic variations by whole-genome sequencing (WGS). This combined genomic approach allowed the identification of three atypical and complex DMD mutations due to a deep intronic variation and two large inversions, respectively. All three mutations affect DMD gene splicing and cause a lack of dystrophin protein production, and one of these also generates unique fusion genes and transcripts. Further characterization of USCs using a novel cell-sorting technology (Celector) highlighted cell-type variability and the representation of cell-specific DMD isoforms. Our comprehensive approach to USCs unraveled RNA, DNA, and cell-specific features and demonstrated that USCs are a robust tool for studying and diagnosing DMD.
Collapse
Affiliation(s)
- Maria S Falzarano
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Andrea Grilli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41121, Italy
| | | | | | - Rachele Rossi
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Francesca Gualandi
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Paola Rimessi
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Reem El Dani
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Marina Fabris
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | | | - Wenyan Li
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | - Elena Pegoraro
- ERN Neuromuscular Center, Department of Neurosciences, Unit of Neurology, University of Padua, Padua 35122, Italy
| | - Luca Bello
- ERN Neuromuscular Center, Department of Neurosciences, Unit of Neurology, University of Padua, Padua 35122, Italy
| | - Andrea Barp
- The NEMO Clinical Center, Neurorehabilitation Unit, University of Milan, Milan 20162, Italy
| | - Valeria A Sansone
- The NEMO Clinical Center, Neurorehabilitation Unit, University of Milan, Milan 20162, Italy
| | - Madhuri Hegde
- PerkinElmer Genomics, 3950 Shackleford Rd., Ste. 195, Duluth, GA 30096, USA
| | - Barbara Roda
- Stem Sel s.r.l., Bologna 40127, Italy
- Department of Chemistry "G. Ciamician," University of Bologna, Bologna 40126, Italy
| | - Pierluigi Reschiglian
- Stem Sel s.r.l., Bologna 40127, Italy
- Department of Chemistry "G. Ciamician," University of Bologna, Bologna 40126, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41121, Italy
| | - Rita Selvatici
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Alessandra Ferlini
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| |
Collapse
|
29
|
The variability in neurological deficits in Duchenne muscular dystrophy patients may be explained by differences in dystrophin glycoprotein complexes in the brain and muscle. Neuroreport 2021; 32:1229-1233. [PMID: 34494993 DOI: 10.1097/wnr.0000000000001710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic neuromuscular disorder. The variability in neurologic deficits in DMD patients may be explained by the fact that (1) dystrophin containing complexes in the brain are more stable than dystrophin containing complexes in the muscle (2) neurons are not affected by the same stresses as muscle and (3) neurons have a greater capacity to buffer increases in intracellular calcium levels. In the muscle, the loss of dystrophin and subsequent loss of dystrophin-associated proteins (DAPs) affects the stability of the dystrophin-glycoprotein complex and calcium ion channels. It causes the sarcolemma of the muscle to tear and calcium ion leak. The subsequent calcium influx leads to calcium dependant proteolysis. In the brain, the structure of the dystrophin-containing complexes is completely different from the muscle. There are several dystrophin isoforms that combine with a completely different set of proteins compared to the muscle to form several different dystrophin-containing complexes. In addition, the loss of dystrophin does not affect the expression of DAPs. The heterogeneity of dystrophin-containing complexes and the continued expression of DAPs will result in more stable dystrophin-containing complexes in the DMD brain. Muscles are under more stress than neurons as they undergo contractions. This combined with txhe fact that the neurons have a better ability to buffer increases in calcium would suggest that neurons are less likely to be damaged despite the loss of dystrophin.
Collapse
|
30
|
Saoudi A, Zarrouki F, Sebrié C, Izabelle C, Goyenvalle A, Vaillend C. Emotional behavior and brain anatomy of the mdx52 mouse model of Duchenne muscular dystrophy. Dis Model Mech 2021; 14:dmm049028. [PMID: 34546327 PMCID: PMC8476816 DOI: 10.1242/dmm.049028] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/30/2021] [Indexed: 11/20/2022] Open
Abstract
The exon-52-deleted mdx52 mouse is a critical model of Duchenne muscular dystrophy (DMD), as it features a deletion in a hotspot region of the DMD gene, frequently mutated in patients. Deletion of exon 52 impedes expression of several brain dystrophins (Dp427, Dp260 and Dp140), thus providing a key model for studying the cognitive impairment associated with DMD and testing rescuing strategies. Here, using in vivo magnetic resonance imaging and neurohistology, we found no gross brain abnormalities in mdx52 mice, suggesting that the neural dysfunctions in this model are likely at the level of brain cellular functionalities. Then, we investigated emotional behavior and fear learning performance of mdx52 mice compared to mdx mice that only lack Dp427 to focus on behavioral phenotypes that could be used in future comparative preclinical studies. mdx52 mice displayed enhanced anxiety and a severe impairment in learning an amygdala-dependent Pavlovian association. These replicable behavioral outcome measures are reminiscent of the internalizing problems reported in a quarter of DMD patients, and will be useful for preclinical estimation of the efficacy of treatments targeting brain dysfunctions in DMD.
Collapse
Affiliation(s)
- Amel Saoudi
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, 91190, Gif-sur-Yvette, France
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | - Faouzi Zarrouki
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, 91190, Gif-sur-Yvette, France
| | - Catherine Sebrié
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401 Orsay, France
| | - Charlotte Izabelle
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, 91190, Gif-sur-Yvette, France
| | - Aurélie Goyenvalle
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, 91190, Gif-sur-Yvette, France
| |
Collapse
|
31
|
Koo J, Park S, Sung SE, Lee J, Kim DS, Lee J, Lee JR, Kim NS, Lee DY. Altered Gene Expression Profiles in Neural Stem Cells Derived from Duchenne Muscular Dystrophy Patients with Intellectual Disability. Exp Neurobiol 2021; 30:263-274. [PMID: 34483141 PMCID: PMC8424384 DOI: 10.5607/en21008] [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: 04/16/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 11/19/2022] Open
Abstract
Intellectual disability (ID) is a neurodevelopmental disorder defined by below-average intelligence (intelligence quotient of <70) accompanied by adaptive behavior deficits. Defects in the functions of neural stem cells during brain development are closely linked to the pathogenesis of ID. To understand the molecular etiology of ID, we examined neural stem cells from individuals with Duchenne muscular dystrophy (DMD), a genetic disorder in which approximately one-third of the patients exhibit ID. In this study, we generated induced pluripotent stem cells from peripheral blood mononuclear cells from a normal individual and DMD patients with and without ID to identify ID-specific functional and molecular abnormalities. We found defects in neural ectoderm formation in the group of DMD patients with ID. Our transcriptome analysis of patient-derived neural stem cells revealed altered expression of genes related to the hippo signaling pathway and neuroactive ligand-receptor interaction, implicating these in the pathogenesis of ID in patients with DMD.
Collapse
Affiliation(s)
- Jahong Koo
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34141, Korea
| | - Subin Park
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Biochemistry, Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Soo-Eun Sung
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Jeehun Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Dae Soo Kim
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34141, Korea.,Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Jungwoon Lee
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34141, Korea.,Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Jae-Ran Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Nam-Soon Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34141, Korea
| | - Da Yong Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
32
|
Biagi L, Lenzi S, Cipriano E, Fiori S, Bosco P, Cristofani P, Astrea G, Pini A, Cioni G, Mercuri E, Tosetti M, Battini R. Neural substrates of neuropsychological profiles in dystrophynopathies: A pilot study of diffusion tractography imaging. PLoS One 2021; 16:e0250420. [PMID: 33939732 PMCID: PMC8092766 DOI: 10.1371/journal.pone.0250420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/06/2021] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Cognitive difficulties and neuropsychological alterations in Duchenne and Becker muscular dystrophy (DMD, BMD) boys are not yet sufficiently explored, although this topic could have a relevant impact, finding novel biomarkers of disease both at genetics and neuroimaging point of view. The current study aims to: 1) analyze the neuropsychological profile of a group of DMD and BMD boys without cognitive impairment with an assessment of their executive functions; 2) explore the structural connectivity in DMD, BMD, and age-matched controls focusing on cortico-subcortical tracts that connect frontal cortex, basal ganglia, and cerebellum via the thalamus; 3) explore possible correlations between altered structural connectivity and clinical neuropsychological measures. MATERIALS AND METHODS This pilot study included 15 boys (5 DMD subjects, 5 BMD subjects, and 5 age-matched typically developing, TD). They were assessed using a neuropsychological assessment protocol including cognitive and executive functioning assessment and performed a 1.5T MRI brain exam including advance Diffusion Weighted Imaging (DWI) method for tractography. Structural connectivity measurements were extracted along three specific tracts: Cortico-Ponto-Cerebellar Tract (CPCT), Cerebellar-Thalamic Tract (CTT), and Superior Longitudinal Fasciculus (SLF). Cortical-Spinal Tract (CST) was selected for reference, as control tract. RESULTS Regarding intellectual functioning, a major impairment in executive functions compared to the general intellectual functioning was observed both for DMD (mean score = 86.20; SD = 11.54) and for BMD children (mean score = 88; SD = 3.67). Mean FA resulted tendentially always lower in DMD compared to both BMD and TD groups for all the examined tracts. The differences in FA were statistically significant for the right CTT (DMD vs BMD, p = 0.002, and DMD vs TD, p = 0.0015) and the right CPCT (DMD vs TD, p = 0.008). Concerning DMD, significant correlations emerged between FA-R-CTT and intellectual quotients (FIQ, p = 0.044; ρs = 0.821), and executive functions (Denomination Total, p = 0.044, ρs = 0.821; Inhibition Total, p = 0.019, ρs = 0.900). BMD showed a significant correlation between FA-R-CPCT and working memory index (p = 0.007; ρs = 0.949). DISCUSSION AND CONCLUSION In this pilot study, despite the limitation of sample size, the findings support the hypothesis of the involvement of a cerebellar-thalamo-cortical loop for the neuropsychological profile of DMD, as the CTT and the CPCT are involved in the network and the related brain structures are known to be implied in executive functions. Our results suggest that altered WM connectivity and reduced fibre organization in cerebellar tracts, probably due to the lack of dystrophin in the brain, may render less efficient some neuropsychological functions in children affected by dystrophinopathies. The wider multicentric study could help to better establish the role of cerebellar connectivity in neuropsychological profile for dystrophinopathies, identifying possible novel diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Laura Biagi
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Sara Lenzi
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Emilio Cipriano
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
- Department of Physics, University of Pisa, Pisa, Italy
| | - Simona Fiori
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Paolo Bosco
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Paola Cristofani
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Guia Astrea
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Antonella Pini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Giovanni Cioni
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Catholic University and Nemo Center, Policlinico Universitario Gemelli, Rome, Italy
| | - Michela Tosetti
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
33
|
Fortunato F, Rossi R, Falzarano MS, Ferlini A. Innovative Therapeutic Approaches for Duchenne Muscular Dystrophy. J Clin Med 2021; 10:jcm10040820. [PMID: 33671409 PMCID: PMC7922390 DOI: 10.3390/jcm10040820] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common childhood muscular dystrophy affecting ~1:5000 live male births. Following the identification of pathogenic variations in the dystrophin gene in 1986, the underlining genotype/phenotype correlations emerged and the role of the dystrophin protein was elucidated in skeletal, smooth, and cardiac muscles, as well as in the brain. When the dystrophin protein is absent or quantitatively or qualitatively modified, the muscle cannot sustain the stress of repeated contractions. Dystrophin acts as a bridging and anchoring protein between the sarcomere and the sarcolemma, and its absence or reduction leads to severe muscle damage that eventually cannot be repaired, with its ultimate substitution by connective tissue and fat. The advances of an understanding of the molecular pathways affected in DMD have led to the development of many therapeutic strategies that tackle different aspects of disease etiopathogenesis, which have recently led to the first successful approved orphan drugs for this condition. The therapeutic advances in this field have progressed exponentially, with second-generation drugs now entering in clinical trials as gene therapy, potentially providing a further effective approach to the condition.
Collapse
|
34
|
Battini R, Lenzi S, Lucibello S, Chieffo D, Moriconi F, Cristofani P, Bulgheroni S, Cumbo F, Pane M, Baranello G, Alfieri P, Astrea G, Cioni G, Vicari S, Mercuri E. Longitudinal data of neuropsychological profile in a cohort of Duchenne muscular dystrophy boys without cognitive impairment. Neuromuscul Disord 2021; 31:319-327. [PMID: 33658162 DOI: 10.1016/j.nmd.2021.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 12/18/2022]
Abstract
The aim of the study was to re-assess neuropsychological profile in a group of boys with Duchenne muscular dystrophy without intellectual disability and neuropsychiatric disorder three years apart from a previous evaluation, to establish possible changes over time. We were also interested in defining more in detail correlation between genotype and neuropsychological phenotype. Thirty-three of the previous 40 subjects (mean age at follow up: 10 years and 7 months) agreed to participate in the follow up study and to perform the new assessment. The results confirm a typical neuropsychological profile, with difficulty in the manipulation of stored information, poor abstract reasoning and planning capacity and impulsiveness, supporting the involvement of a cerebellar striatal cortical network for these children. The more detailed description of subgroups of subjects, according to the real expression of Dp140, let to reveal possible genotype-neuropsychological phenotype correlations, and a more general neuropsychological impairment emerged in boys without Dp140 expression.
Collapse
Affiliation(s)
- R Battini
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 341/ ABC, Calambrone, Pisa 56128, Italy; Department of Clinical and Experimental Medicine, University of Pisa, Via Savi10, 56126 Pisa, Italy.
| | - S Lenzi
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 341/ ABC, Calambrone, Pisa 56128, Italy
| | - S Lucibello
- Pediatric Neurology Unit, Catholic University and Nemo Center, Policlinico Universitario Gemelli Largo Agostino Gemelli, 8. Rome, Italy
| | - D Chieffo
- Pediatric Neurology Unit, Catholic University and Nemo Center, Policlinico Universitario Gemelli Largo Agostino Gemelli, 8. Rome, Italy
| | - F Moriconi
- Pediatric Neurology Unit, Catholic University and Nemo Center, Policlinico Universitario Gemelli Largo Agostino Gemelli, 8. Rome, Italy
| | - P Cristofani
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 341/ ABC, Calambrone, Pisa 56128, Italy
| | - S Bulgheroni
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - F Cumbo
- Child and Adolescent Neuropsychiatry Unit, Department of Neurological and Psychiatric Science, IRCCS Bambino Gesù Children's Hospital, Piazza di Sant'Onofrio, 4. Rome 00165, Italy
| | - M Pane
- Pediatric Neurology Unit, Catholic University and Nemo Center, Policlinico Universitario Gemelli Largo Agostino Gemelli, 8. Rome, Italy
| | - G Baranello
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - P Alfieri
- Child and Adolescent Neuropsychiatry Unit, Department of Neurological and Psychiatric Science, IRCCS Bambino Gesù Children's Hospital, Piazza di Sant'Onofrio, 4. Rome 00165, Italy
| | - G Astrea
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 341/ ABC, Calambrone, Pisa 56128, Italy
| | - G Cioni
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 341/ ABC, Calambrone, Pisa 56128, Italy; Department of Clinical and Experimental Medicine, University of Pisa, Via Savi10, 56126 Pisa, Italy
| | - S Vicari
- Child and Adolescent Neuropsychiatry Unit, Department of Neurological and Psychiatric Science, IRCCS Bambino Gesù Children's Hospital, Piazza di Sant'Onofrio, 4. Rome 00165, Italy; Department of Life Sciences and Public Health, Catholic University, Largo Agostino Gemelli, 8, Rome, Italy
| | - E Mercuri
- Pediatric Neurology Unit, Catholic University and Nemo Center, Policlinico Universitario Gemelli Largo Agostino Gemelli, 8. Rome, Italy
| |
Collapse
|
35
|
Belmaati Cherkaoui M, Vacca O, Izabelle C, Boulay AC, Boulogne C, Gillet C, Barnier JV, Rendon A, Cohen-Salmon M, Vaillend C. Dp71 contribution to the molecular scaffold anchoring aquaporine-4 channels in brain macroglial cells. Glia 2020; 69:954-970. [PMID: 33247858 DOI: 10.1002/glia.23941] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
Intellectual disability in Duchenne muscular dystrophy has been associated with the loss of dystrophin-protein 71, Dp71, the main dystrophin-gene product in the adult brain. Dp71 shows major expression in perivascular macroglial endfeet, suggesting that dysfunctional glial mechanisms contribute to cognitive impairments. In the present study, we investigated the molecular alterations induced by a selective loss of Dp71 in mice, using semi-quantitative immunogold analyses in electron microscopy and immunofluorescence confocal analyses in brain sections and purified gliovascular units. In macroglial pericapillary endfeet of the cerebellum and hippocampus, we found a drastic reduction (70%) of the polarized distribution of aquaporin-4 (AQP4) channels, a 50% reduction of β-dystroglycan, and a complete loss of α1-syntrophin. Interestingly, in the hippocampus and cortex, these effects were not homogeneous: AQP4 and AQP4ex isoforms were mostly lost around capillaries but preserved in large vessels corresponding to pial arteries, penetrating cortical arterioles, and arterioles of the hippocampal fissure, indicating the presence of Dp71-independent pools of AQP4 in these vascular structures. In conclusion, the depletion of Dp71 strongly alters the distribution of AQP4 selectively in macroglial perivascular endfeet surrounding capillaries. This effect likely affects water homeostasis and blood-brain barrier functions and may thus contribute to the synaptic and cognitive defects associated with Dp71 deficiency.
Collapse
Affiliation(s)
| | - Ophélie Vacca
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, Gif-sur-Yvette, France
| | - Charlotte Izabelle
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, Gif-sur-Yvette, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Unité Mixte de Recherche 7241CNRS, Unité 1050 INSERM, PSL Research University, Paris, France
| | - Claire Boulogne
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Cynthia Gillet
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Jean-Vianney Barnier
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, Gif-sur-Yvette, France
| | - Alvaro Rendon
- UPMC Université Paris 06, INSERM, CNRS, Institut de la Vision, Sorbonne Universités, Paris, France
| | - Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Unité Mixte de Recherche 7241CNRS, Unité 1050 INSERM, PSL Research University, Paris, France
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris Saclay, Gif-sur-Yvette, France
| |
Collapse
|
36
|
Yablonka-Reuveni Z, Stockdale F, Nudel U, Israeli D, Blau HM, Shainberg A, Neuman S, Kessler-Icekson G, Krull EM, Paterson B, Fuchs OS, Greenberg D, Sarig R, Halevy O, Ozawa E, Katcoff DJ. Farewell to Professor David Yaffe - A pillar of the myogenesis field. Eur J Transl Myol 2020; 30:9306. [PMID: 33117511 PMCID: PMC7582454 DOI: 10.4081/ejtm.2020.9306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
It is with great sadness that we have learned about the passing of Professor David Yaffe (1929-2020, Israel). Yehi Zichro Baruch - May his memory be a blessing. David was a man of family, science and nature. A native of Israel, David grew up in the historic years that preceded the birth of the State of Israel. He was a member of the group that established Kibbutz Revivim in the Negev desert, and in 1948 participated in Israel's War of Independence. David and Ruth eventually joined Kibbutz Givat Brenner by Rehovot, permitting David to be both a kibbutz member and a life-long researcher at the Weizmann Institute of Science, where David received his PhD in 1959. David returned to the Institute after his postdoc at Stanford. Here, after several years of researching a number of tissues as models for studying the process of differentiation, David entered the myogenesis field and stayed with it to his last day. With his dedication to the field of myogenesis and his commitment to furthering the understanding of the People and the Land of Israel throughout the international scientific community, David organized the first ever myogenesis meeting that took place in Shoresh, Israel in 1975. This was followed by the 1980 myogenesis meeting at the same place and many more outstanding meetings, all of which brought together myogenesis, nature and scenery. Herein, through the preparation and publication of this current manuscript, we are meeting once again at a "David Yaffe myogenesis meeting". Some of us have been members of the Yaffe lab, some of us have known David as his national and international colleagues in the myology field. One of our contributors has also known (and communicates here) about David Yaffe's earlier years as a kibbutznick in the Negev. Our collective reflections are a tribute to Professor David Yaffe. We are fortunate that the European Journal of Translational Myology has provided us with tremendous input and a platform for holding this 2020 distance meeting "Farwell to Professor David Yaffe - A Pillar of the Myogenesis Field".
Collapse
Affiliation(s)
- Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Uri Nudel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Helen M. Blau
- Stanford University School of Medicine, Institute for Stem Cell Biology and Regenerative Medicine, Department of Microbiology and Immunology, Clinical Sciences Research Center, Stanford, CA, USA
| | - Asher Shainberg
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Gania Kessler-Icekson
- Laboratory of Cellular and Molecular Cardiology, Felsenstein Medical Research Center, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Bruce Paterson
- Laboratory of Biochemistry and Molecular Biology, National Institutes of Health, Bethesda, Maryland, USA
| | | | - David Greenberg
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachel Sarig
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Orna Halevy
- Faculty of Agriculture, The Hebrew University, Rehovot, Israel
| | - Eijiro Ozawa
- National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Don J. Katcoff
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| |
Collapse
|
37
|
Hendriksen JGM, Thangarajh M, Kan HE, Muntoni F. 249th ENMC International Workshop: The role of brain dystrophin in muscular dystrophy: Implications for clinical care and translational research, Hoofddorp, The Netherlands, November 29th-December 1st 2019. Neuromuscul Disord 2020; 30:782-794. [PMID: 32912717 DOI: 10.1016/j.nmd.2020.08.357] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/27/2020] [Accepted: 08/10/2020] [Indexed: 01/16/2023]
Affiliation(s)
- Jos G M Hendriksen
- Kempenhaeghe Center For Neurological Learning Disabilities, Heeze, the Netherlands; Duchenne Center, the Netherlands
| | - Mathula Thangarajh
- Department of Neurology, Virginia Commonwealth University, Richmond, USA
| | - Hermien E Kan
- Duchenne Center, the Netherlands; C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
38
|
Romo-Yáñez J, Rodríguez-Martínez G, Aragón J, Siqueiros-Márquez L, Herrera-Salazar A, Velasco I, Montanez C. Characterization of the expression of dystrophins and dystrophin-associated proteins during embryonic neural stem/progenitor cell differentiation. Neurosci Lett 2020; 736:135247. [DOI: 10.1016/j.neulet.2020.135247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 07/05/2020] [Accepted: 07/10/2020] [Indexed: 10/23/2022]
|
39
|
Brignol TN, Mornet D, Montañez C, Ventura DF, Vaillend C, Rendón A. What is the ocular phenotype associated with a single exon 78 deletion in Duchenne muscular dystrophy? J Hum Genet 2020; 65:715-716. [DOI: 10.1038/s10038-020-0755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 03/13/2020] [Indexed: 11/09/2022]
|
40
|
Hildyard JCW, Crawford AH, Rawson F, Riddell DO, Harron RCM, Piercy RJ. Single-transcript multiplex in situ hybridisation reveals unique patterns of dystrophin isoform expression in the developing mammalian embryo. Wellcome Open Res 2020; 5:76. [PMID: 32724863 PMCID: PMC7372313 DOI: 10.12688/wellcomeopenres.15762.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2020] [Indexed: 12/24/2022] Open
Abstract
Background: The dystrophin gene has multiple isoforms: full-length dystrophin (dp427) is principally known for its expression in skeletal and cardiac muscle, but is also expressed in the brain, and several internal promoters give rise to shorter, N-terminally truncated isoforms with wider tissue expression patterns (dp260 in the retina, dp140 in the brain and dp71 in many tissues). These isoforms are believed to play unique cellular roles both during embryogenesis and in adulthood, but their shared sequence identity at both mRNA and protein levels makes study of distinct isoforms challenging by conventional methods. Methods: RNAscope is a novel in-situ hybridisation technique that offers single-transcript resolution and the ability to multiplex, with different target sequences assigned to distinct fluorophores. Using probes designed to different regions of the dystrophin transcript (targeting 5', central and 3' sequences of the long dp427 mRNA), we can simultaneously detect and distinguish multiple dystrophin mRNA isoforms at sub-cellular histological levels. We have used these probes in healthy and dystrophic canine embryos to gain unique insights into isoform expression and distribution in the developing mammal. Results: Dp427 is found in developing muscle as expected, apparently enriched at nascent myotendinous junctions. Endothelial and epithelial surfaces express dp71 only. Within the brain and spinal cord, all three isoforms are expressed in spatially distinct regions: dp71 predominates within proliferating germinal layer cells, dp140 within maturing, migrating cells and dp427 appears within more established cell populations. Dystrophin is also found within developing bones and teeth, something previously unreported, and our data suggests orchestrated involvement of multiple isoforms in formation of these tissues. Conclusions: Overall, shorter isoforms appear associated with proliferation and migration, and longer isoforms with terminal lineage commitment: we discuss the distinct structural contributions and transcriptional demands suggested by these findings.
Collapse
Affiliation(s)
- John C. W. Hildyard
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Abbe H. Crawford
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Faye Rawson
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Dominique O. Riddell
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Rachel C. M. Harron
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Richard J. Piercy
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| |
Collapse
|
41
|
Doorenweerd N. Combining genetics, neuropsychology and neuroimaging to improve understanding of brain involvement in Duchenne muscular dystrophy - a narrative review. Neuromuscul Disord 2020; 30:437-442. [PMID: 32522501 DOI: 10.1016/j.nmd.2020.05.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 11/24/2022]
Abstract
Duchenne muscular dystrophy is a multifactorial disease including a cognitive phenotype. It is caused by mutations in the X-chromosomal DMD gene from which dystrophin is synthesized. Multiple isoforms of dystrophin have been identified. The full length dystrophin isoform Dp427m is expressed predominantly in muscle. Other isoforms include: Dp427c, Dp427p, Dp260, Dp140, Dp116, Dp71 and Dp40. The majority of these isoforms are expressed in brain and several hypotheses exist on their role in subtypes of neurons and astrocytes. However, their function in relation to cognition remains unclear. Unlike progressive muscle wasting, cognitive involvement is not seen in all DMD patients and the severity varies greatly. To achieve a better understanding of brain involvement in DMD, a multidisciplinary approach is required. Here, we review the latest findings on dystrophin isoform expression in the brain; specific DMD-associated learning and behavioural difficulties; and imaging and spectroscopy findings relating to brain structure, networks, perfusion and metabolism. The main challenge lies in determining links between these different findings. If we can determine which factors play a role in the differentiation between severe and minor cognitive problems in DMD in the near future, we can both provide better advise for the patients and also develop targeted therapeutic interventions.
Collapse
Affiliation(s)
- Nathalie Doorenweerd
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, C-03-Q, P.O. Box 9600, 2300 RC Leiden, The Netherlands; John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom.
| |
Collapse
|
42
|
Hildyard JCW, Crawford AH, Rawson F, Riddell DO, Harron RCM, Piercy RJ. Single-transcript multiplex in situ hybridisation reveals unique patterns of dystrophin isoform expression in the developing mammalian embryo. Wellcome Open Res 2020; 5:76. [PMID: 32724863 PMCID: PMC7372313 DOI: 10.12688/wellcomeopenres.15762.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2020] [Indexed: 07/30/2023] Open
Abstract
Background: The dystrophin gene has multiple isoforms: full-length dystrophin (dp427) is principally known for its expression in skeletal and cardiac muscle, but is also expressed in the brain, and several internal promoters give rise to shorter, N-terminally truncated isoforms with wider tissue expression patterns (dp260 in the retina, dp140 in the brain and dp71 in many tissues). These isoforms are believed to play unique cellular roles both during embryogenesis and in adulthood, but their shared sequence identity at both mRNA and protein levels makes study of distinct isoforms challenging by conventional methods. Methods: RNAscope is a novel in-situ hybridisation technique that offers single-transcript resolution and the ability to multiplex, with different target sequences assigned to distinct fluorophores. Using probes designed to different regions of the dystrophin transcript (targeting 5', central and 3' sequences of the long dp427 mRNA), we can simultaneously detect and distinguish multiple dystrophin mRNA isoforms at sub-cellular histological levels. We have used these probes in healthy and dystrophic canine embryos to gain unique insights into isoform expression and distribution in the developing mammal. Results: Dp427 is found in developing muscle as expected, apparently enriched at nascent myotendinous junctions. Endothelial and epithelial surfaces express dp71 only. Within the brain and spinal cord, all three isoforms are expressed in spatially distinct regions: dp71 predominates within proliferating germinal layer cells, dp140 within maturing, migrating cells and dp427 appears within more established cell populations. Dystrophin is also found within developing bones and teeth, something previously unreported, and our data suggests orchestrated involvement of multiple isoforms in formation of these tissues. Conclusions: Overall, shorter isoforms appear associated with proliferation and migration, and longer isoforms with terminal lineage commitment: we discuss the distinct structural contributions and transcriptional demands suggested by these findings.
Collapse
Affiliation(s)
- John C. W. Hildyard
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Abbe H. Crawford
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Faye Rawson
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Dominique O. Riddell
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Rachel C. M. Harron
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| | - Richard J. Piercy
- Department of Clinical Science and Services, Royal Veterinary College, London, Camden, London, NW1 0TU, UK
| |
Collapse
|
43
|
Barboni MTS, Vaillend C, Joachimsthaler A, Liber AMP, Khabou H, Roux MJ, Vacca O, Vignaud L, Dalkara D, Guillonneau X, Ventura DF, Rendon A, Kremers J. Rescue of Defective Electroretinographic Responses in Dp71-Null Mice With AAV-Mediated Reexpression of Dp71. Invest Ophthalmol Vis Sci 2020; 61:11. [PMID: 32049345 PMCID: PMC7326481 DOI: 10.1167/iovs.61.2.11] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To study the potential effect of a gene therapy, designed to rescue the expression of dystrophin Dp71 in the retinas of Dp71-null mice, on retinal physiology. Methods We recorded electroretinograms (ERGs) in Dp71-null and wild-type littermate mice. In dark-adapted eyes, responses to flashes of several strengths were measured. In addition, flash responses on a 25-candela/square meters background were measured. On- and Off-mediated responses to sawtooth stimuli and responses to photopic sine-wave modulation (3–30 Hz) were also recorded. After establishing the ERG phenotype, the ShH10-GFP adeno-associated virus (AAV), which has been previously shown to target specifically Müller glial cells (MGCs), was delivered intravitreously with or without (sham therapy) the Dp71 coding sequence under control of a CBA promoter. ERG recordings were repeated three months after treatment. Real-time quantitative PCR and Western blotting analyses were performed in order to quantify Dp71 expression in the retinas. Results Dp71-null mice displayed reduced b-waves in dark- and light-adapted flash ERGs and smaller response amplitudes to photopic rapid-on sawtooth modulation and to sine-wave stimuli. Three months after intravitreal injections of the ShH10-GFP-2A-Dp71 AAV vector, ERG responses were completely recovered in treated eyes of Dp71-null mice. The functional rescue was associated with an overexpression of Dp71 in treated retinas. Conclusions The present results show successful functional recovery accompanying the reexpression of Dp71. In addition, this experimental model sheds light on MGCs influencing ERG components, since previous reports showed that aquaporin 4 and Kir4.1 channels were mislocated in MGCs of Dp71-null mice, while their distribution could be normalized following intravitreal delivery of the same ShH10-GFP-2A-Dp71 vector.
Collapse
|
44
|
Naidoo M, Anthony K. Dystrophin Dp71 and the Neuropathophysiology of Duchenne Muscular Dystrophy. Mol Neurobiol 2020; 57:1748-1767. [PMID: 31836945 PMCID: PMC7060961 DOI: 10.1007/s12035-019-01845-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
Duchenne muscular dystrophy (DMD) is caused by frameshift mutations in the DMD gene that prevent the body-wide translation of its protein product, dystrophin. Besides a severe muscle phenotype, cognitive impairment and neuropsychiatric symptoms are prevalent. Dystrophin protein 71 (Dp71) is the major DMD gene product expressed in the brain and mutations affecting its expression are associated with the DMD neuropsychiatric syndrome. As with dystrophin in muscle, Dp71 localises to dystrophin-associated protein complexes in the brain. However, unlike in skeletal muscle; in the brain, Dp71 is alternatively spliced to produce many isoforms with differential subcellular localisations and diverse cellular functions. These include neuronal differentiation, adhesion, cell division and excitatory synapse organisation as well as nuclear functions such as nuclear scaffolding and DNA repair. In this review, we first describe brain involvement in DMD and the abnormalities observed in the DMD brain. We then review the gene expression, RNA processing and functions of Dp71. We review genotype-phenotype correlations and discuss emerging cellular/tissue evidence for the involvement of Dp71 in the neuropathophysiology of DMD. The literature suggests changes observed in the DMD brain are neurodevelopmental in origin and that their risk and severity is associated with a cumulative loss of distal DMD gene products such as Dp71. The high risk of neuropsychiatric syndromes in Duchenne patients warrants early intervention to achieve the best possible quality of life. Unravelling the function and pathophysiological significance of dystrophin in the brain has become a high research priority to inform the development of brain-targeting treatments for Duchenne.
Collapse
Affiliation(s)
- Michael Naidoo
- Centre for Physical Activity and Life Sciences, Faculty of Arts, Science and Technology, University of Northampton, University Drive, Northampton, Northamptonshire, NN1 5PH, UK
| | - Karen Anthony
- Centre for Physical Activity and Life Sciences, Faculty of Arts, Science and Technology, University of Northampton, University Drive, Northampton, Northamptonshire, NN1 5PH, UK.
| |
Collapse
|
45
|
Patel AM, Wierda K, Thorrez L, van Putten M, De Smedt J, Ribeiro L, Tricot T, Gajjar M, Duelen R, Van Damme P, De Waele L, Goemans N, Tanganyika-de Winter C, Costamagna D, Aartsma-Rus A, van Duyvenvoorde H, Sampaolesi M, Buyse GM, Verfaillie CM. Dystrophin deficiency leads to dysfunctional glutamate clearance in iPSC derived astrocytes. Transl Psychiatry 2019; 9:200. [PMID: 31434868 PMCID: PMC6704264 DOI: 10.1038/s41398-019-0535-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) results, beside muscle degeneration in cognitive defects. As neuronal function is supported by astrocytes, which express dystrophin, we hypothesized that loss of dystrophin from DMD astrocytes might contribute to these cognitive defects. We generated cortical neuronal and astrocytic progeny from induced pluripotent stem cells (PSC) from six DMD subjects carrying different mutations and several unaffected PSC lines. DMD astrocytes displayed cytoskeletal abnormalities, defects in Ca+2 homeostasis and nitric oxide signaling. In addition, defects in glutamate clearance were identified in DMD PSC-derived astrocytes; these deficits were related to a decreased neurite outgrowth and hyperexcitability of neurons derived from healthy PSC. Read-through molecule restored dystrophin expression in DMD PSC-derived astrocytes harboring a premature stop codon mutation, corrected the defective astrocyte glutamate clearance and prevented associated neurotoxicity. We propose a role for dystrophin deficiency in defective astroglial glutamate homeostasis which initiates defects in neuronal development.
Collapse
Affiliation(s)
- Abdulsamie M. Patel
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Keimpe Wierda
- 0000000104788040grid.11486.3aCenter for Brain & Disease Research, VIB, Leuven, Belgium
| | - Lieven Thorrez
- 0000 0001 0668 7884grid.5596.fKU Leuven Department of Development and Regeneration, Campus Kulak, Kortrijk, Belgium
| | - Maaike van Putten
- 0000000089452978grid.10419.3dDepartment of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jonathan De Smedt
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Luis Ribeiro
- 0000000104788040grid.11486.3aCenter for Brain & Disease Research, VIB, Leuven, Belgium
| | - Tine Tricot
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Madhavsai Gajjar
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Robin Duelen
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium ,0000 0001 0668 7884grid.5596.fTranslational Cardiomyology Lab, Stem Cell Biology and Embryology Unit, KU Leuven, Leuven, Belgium
| | - Philip Van Damme
- 0000000104788040grid.11486.3aCenter for Brain & Disease Research, VIB, Leuven, Belgium ,0000 0001 0668 7884grid.5596.fLaboratory of Neurobiology, Department of Neuroscience, KU Leuven, Leuven, Belgium ,0000 0004 0626 3338grid.410569.fNeurology Department, University Hospitals Leuven, Leuven, Belgium
| | - Liesbeth De Waele
- 0000 0001 0668 7884grid.5596.fKU Leuven Department of Development and Regeneration, Campus Kulak, Kortrijk, Belgium ,0000 0004 0626 3338grid.410569.fDepartment of Paediatric Child Neurology, University Hospitals Leuven, Leuven, Belgium ,0000 0001 0668 7884grid.5596.fVesalius Research Center, Laboratory of Neurobiology, KU Leuven, Leuven, Belgium
| | - Nathalie Goemans
- 0000 0004 0626 3338grid.410569.fDepartment of Paediatric Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Christa Tanganyika-de Winter
- 0000000089452978grid.10419.3dDepartment of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Domiziana Costamagna
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium ,0000 0001 0668 7884grid.5596.fTranslational Cardiomyology Lab, Stem Cell Biology and Embryology Unit, KU Leuven, Leuven, Belgium
| | - Annemieke Aartsma-Rus
- 0000000089452978grid.10419.3dDepartment of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermine van Duyvenvoorde
- 0000000089452978grid.10419.3dLaboratory for Diagnostic Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands
| | - Maurilio Sampaolesi
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium ,0000 0001 0668 7884grid.5596.fTranslational Cardiomyology Lab, Stem Cell Biology and Embryology Unit, KU Leuven, Leuven, Belgium
| | - Gunnar M. Buyse
- 0000 0004 0626 3338grid.410569.fDepartment of Paediatric Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Catherine M. Verfaillie
- 0000 0001 0668 7884grid.5596.fStem Cell Institute Leuven, Dept. of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
46
|
Dystrobrevin is required postsynaptically for homeostatic potentiation at the Drosophila NMJ. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1579-1591. [PMID: 30904609 DOI: 10.1016/j.bbadis.2019.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 11/20/2022]
Abstract
Evolutionarily conserved homeostatic systems have been shown to modulate synaptic efficiency at the neuromuscular junctions of organisms. While advances have been made in identifying molecules that function presynaptically during homeostasis, limited information is currently available on how postsynaptic alterations affect presynaptic function. We previously identified a role for postsynaptic Dystrophin in the maintenance of evoked neurotransmitter release. We herein demonstrated that Dystrobrevin, a member of the Dystrophin Glycoprotein Complex, was delocalized from the postsynaptic region in the absence of Dystrophin. A newly-generated Dystrobrevin mutant showed elevated evoked neurotransmitter release, increased bouton numbers, and a readily releasable pool of synaptic vesicles without changes in the function or numbers of postsynaptic glutamate receptors. In addition, we provide evidence to show that the highly conserved Cdc42 Rho GTPase plays a key role in the postsynaptic Dystrophin/Dystrobrevin pathway for synaptic homeostasis. The present results give novel insights into the synaptic deficits underlying Duchenne Muscular Dystrophy affected by a dysfunctional Dystrophin Glycoprotein complex.
Collapse
|
47
|
Abstract
A new concept of pathogenesis Duchenne Muscular Dystrophy (DMD) based on authors experience and discussion of results of other studies. Author estimate DMD as a cerebro-muscular pathology depend on absent dystrophin (D) in skeletal muscles, heart, brain. Author suppose that all family D work as one functional system, which play the important role in organization of walking. Destroy of this system is cause of the disease.
Collapse
Affiliation(s)
- L P Grinio
- Evdokimov State Moscow University of Medical Dentistry, Moscow, Russia
| |
Collapse
|
48
|
Jantrapirom S, Cao DS, Wang JW, Hing H, Tabone CJ, Lantz K, de Belle JS, Qiu YT, Smid HM, Yamaguchi M, Fradkin LG, Noordermeer JN, Potikanond S. Dystrophin is required for normal synaptic gain in the Drosophila olfactory circuit. Brain Res 2019; 1712:158-166. [PMID: 30711401 DOI: 10.1016/j.brainres.2019.01.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 02/03/2023]
Abstract
The Drosophila olfactory system provides an excellent model to elucidate the neural circuits that control behaviors elicited by environmental stimuli. Despite significant progress in defining olfactory circuit components and their connectivity, little is known about the mechanisms that transfer the information from the primary antennal olfactory receptor neurons to the higher order brain centers. Here, we show that the Dystrophin Dp186 isoform is required in the olfactory system circuit for olfactory functions. Using two-photon calcium imaging, we found the reduction of calcium influx in olfactory receptor neurons (ORNs) and also the defect of GABAA mediated inhibitory input in the projection neurons (PNs) in Dp186 mutation. Moreover, the Dp186 mutant flies which display a decreased odor avoidance behavior were rescued by Dp186 restoration in the Drosophila olfactory neurons in either the presynaptic ORNs or the postsynaptic PNs. Therefore, these results revealed a role for Dystrophin, Dp 186 isoform in gain control of the olfactory synapse via the modulation of excitatory and inhibitory synaptic inputs to olfactory projection neurons.
Collapse
Affiliation(s)
- Salinee Jantrapirom
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Thailand
| | - De-Shou Cao
- Division of Biological Sciences, University of California-San Diego, La Jolla, CA 92093, USA
| | - Jing W Wang
- Division of Biological Sciences, University of California-San Diego, La Jolla, CA 92093, USA
| | - Huey Hing
- Department of Biology, State University of New York, Brockport, NY, USA
| | | | - Kathryn Lantz
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA
| | | | - Yu Tong Qiu
- Laboratory of Entomology, Wageningen University, Wageningen, The Netherlands
| | - Hans M Smid
- Laboratory of Entomology, Wageningen University, Wageningen, The Netherlands
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo, Kyoto 606-8585, Japan
| | - Lee G Fradkin
- Laboratory of Developmental Neurobiology, Department of Molecular and Cell Biology, Leiden University Medical Center, Leiden, The Netherlands; University of Massachusetts Medical School, MA, USA
| | - Jasprina N Noordermeer
- Laboratory of Developmental Neurobiology, Department of Molecular and Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Thailand; Laboratory of Developmental Neurobiology, Department of Molecular and Cell Biology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
49
|
Ruggieri S, Viggiano L, Annese T, Rubolino C, Gerbino A, De Zio R, Corsi P, Tamma R, Ribatti D, Errede M, Operto F, Margari L, Resta N, Di Tommaso S, Rosati J, Trojano M, Nico B. DP71 and SERCA2 alteration in human neurons of a Duchenne muscular dystrophy patient. Stem Cell Res Ther 2019; 10:29. [PMID: 30646960 PMCID: PMC6334379 DOI: 10.1186/s13287-018-1125-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/27/2018] [Accepted: 12/25/2018] [Indexed: 01/16/2023] Open
Abstract
Cognitive deficit has been identified in one third of patients affected by Duchenne Muscular Dystrophy, primarily attributed to loss of the short Dp71 dystrophin, the major brain dystrophin isoform. In this study, we investigated for the first time the Dp71 and Dp71-associated proteins cellular localization and expression in human neurons obtained by differentiation from induced pluripotent stem cell line of a patient affected by cognitive impairment. We found structural and molecular alterations in both pluripotent stem cell and derived neurons, reduced Dp71 expression, and a Ca2+ cytoplasmic overload in neurons coupled with increased expression of the SERCA2 pump in the dystrophic neurons. These results suggest that the reduction of Dp71 protein in the Duchenne muscular dystrophy neurons leads to alterations in SERCA2 and to elevated cytosolic Ca2+ concentration with consequent potential disruption of the dystrophin proteins and Dp71-associated proteins.
Collapse
Affiliation(s)
- Simona Ruggieri
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | | | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | | | - Andrea Gerbino
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| | - Roberta De Zio
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| | - Patrizia Corsi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Mariella Errede
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Francesca Operto
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Lucia Margari
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Bari, Italy
| | - Nicoletta Resta
- Division of Medical Genetics, Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari Medical School, Bari, Italy
| | - Silvia Di Tommaso
- Division of Medical Genetics, Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari Medical School, Bari, Italy
| | - Jessica Rosati
- Cellular Reprogramming Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Maria Trojano
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Beatrice Nico
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
50
|
Hoogland G, Hendriksen RGF, Slegers RJ, Hendriks MPH, Schijns OEMG, Aalbers MW, Vles JSH. The expression of the distal dystrophin isoforms Dp140 and Dp71 in the human epileptic hippocampus in relation to cognitive functioning. Hippocampus 2018; 29:102-110. [PMID: 30069964 DOI: 10.1002/hipo.23015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/11/2018] [Accepted: 07/12/2018] [Indexed: 01/06/2023]
Abstract
Dystrophin is an important protein within the central nervous system. The absence of dystrophin, characterizing Duchenne muscular dystrophy (DMD), is associated with brain related comorbidities such as neurodevelopmental (e.g., cognitive and behavioural) deficits and epilepsy. Especially mutations in the downstream part of the DMD gene affecting the dystrophin isoforms Dp140 and Dp71 are found to be associated with cognitive deficits. However, the function of Dp140 is currently not well understood and its expression pattern has previously been implicated to be developmentally regulated. Therefore, we evaluated Dp140 and Dp71 expression in human hippocampi in relation to cognitive functioning in patients with drug-resistant temporal lobe epilepsy (TLE) and post-mortem controls. Hippocampal samples obtained as part of epilepsy surgery were quantitatively analyzed by Western blot and correlations with neuropsychological test results (i.e., memory and intelligence) were examined. First, we demonstrated that the expression of Dp140 does not appear to differ across different ages throughout adulthood. Second, we identified an inverse correlation between memory loss (i.e., verbal and visual memory), but not intelligence (i.e., neither verbal nor performance), and hippocampal Dp140 expression. Finally, patients with TLE appeared to have similar Dp140 expression levels compared to post-mortem controls without neurological disease. Dp140 may thus have a function in normal cognitive (i.e., episodic memory) processes.
Collapse
Affiliation(s)
- Govert Hoogland
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ruben G F Hendriksen
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Rutger J Slegers
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marc P H Hendriks
- Kempenhaeghe Epilepsy Centre, Heeze, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Olaf E M G Schijns
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marlien W Aalbers
- Department of Neurosurgery, Groningen University Medical Centre, Groningen, The Netherlands
| | - Johan S H Vles
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|