1
|
Geuens S, Van Dessel J, Kan HE, Govaarts R, Niks EH, Goemans N, Lemiere J, Doorenweerd N, De Waele L. Genotype and corticosteroid treatment are distinctively associated with gray matter characteristics in patients with Duchenne muscular dystrophy. Neuromuscul Disord 2024; 45:105238. [PMID: 39522443 DOI: 10.1016/j.nmd.2024.105238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
This study investigated if structural variation in specific gray matter areas is associated with corticosteroid treatment or genotype, and if cerebral morphological variations are related to neuropsychological and behavioral outcomes. The CAT12 toolbox in SPM was used for MRI segmentations, assessing subcortical structures, cortical thickness, gyrification, and sulci depths for DMD patients (n = 40; 9-18 years) and age-matched controls (n = 40). Comparisons were made between DMD vs. controls, daily vs. intermittent corticosteroid treatment (n = 20 each), and Dp140+ vs. Dp140- gene mutations (n = 15 vs. 25). MANCOVA, CAT12 3D statistics and Pearson correlations were conducted. DMD patients showed differences in volumes of distinct subcortical structures, left hemisphere cortical thickness, and gyrification in multiple brain areas compared with healthy controls. The daily treated DMD group exhibited differences in subcortical volumes and different patterns of cortical thickness, sulci depth, and gyrification compared to the intermittent treated DMD group. DMD Dp140+ patients displayed altered gyrification and sulci depth compared to DMD Dp140- patients. Finally, we found correlations between neurobehavioral outcomes and brain areas that showed differences in cortical morphology associated with corticosteroid treatment. Both genotype and corticosteroid treatment are associated with variations in subcortical volumes and cortical morphology, albeit in different ways. Corticosteroid treatment appears to have a more profound association with differences in gray matter characteristics of brain regions that are associated with functional outcomes.
Collapse
Affiliation(s)
- Sam Geuens
- University Hospitals Leuven, Child Neurology, Leuven, Belgium; KU Leuven, Department of Development and Regeneration, Leuven, Belgium.
| | - Jeroen Van Dessel
- Center for Developmental Psychiatry, Department of Neurosciences, UPC-KU Leuven, Belgium
| | - Hermien E Kan
- Leiden University Medical Center, C.J. Gorter MRI Center, Department of Radiology, Netherlands; Duchenne Center Netherlands
| | - Rosanne Govaarts
- Leiden University Medical Center, C.J. Gorter MRI Center, Department of Radiology, Netherlands; Duchenne Center Netherlands
| | - Erik H Niks
- Duchenne Center Netherlands; Leiden University Medical Center, Department of Neurology, Netherlands
| | | | - Jurgen Lemiere
- University Hospitals Leuven, Pediatric Hemato-Oncology, Belgium; KU Leuven, Department Oncology, Pediatric Oncology, Belgium
| | - Nathalie Doorenweerd
- Leiden University Medical Center, C.J. Gorter MRI Center, Department of Radiology, Netherlands
| | - Liesbeth De Waele
- University Hospitals Leuven, Child Neurology, Leuven, Belgium; KU Leuven, Department of Development and Regeneration, Leuven, Belgium
| |
Collapse
|
2
|
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
|
3
|
Prigogine C, Ruiz JM, Cebolla AM, Deconinck N, Servais L, Gailly P, Dan B, Cheron G. Cerebellar dysfunction in the mdx mouse model of Duchenne muscular dystrophy: An electrophysiological and behavioural study. Eur J Neurosci 2024; 60:6470-6489. [PMID: 39415418 DOI: 10.1111/ejn.16566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Patients with Duchenne muscular dystrophy (DMD) commonly show specific cognitive deficits in addition to a severe muscle impairment caused by the absence of dystrophin expression in skeletal muscle. These cognitive deficits have been related to the absence of dystrophin in specific regions of the central nervous system, notably cerebellar Purkinje cells (PCs). Dystrophin has recently been involved in GABAA receptors clustering at postsynaptic densities, and its absence, by disrupting this clustering, leads to decreased inhibitory input to PC. We performed an in vivo electrophysiological study of the dystrophin-deficient muscular dystrophy X-linked (mdx) mouse model of DMD to compare PC firing and local field potential (LFP) in alert mdx and control C57Bl/10 mice. We found that the absence of dystrophin is associated with altered PC firing and the emergence of fast (~160-200 Hz) LFP oscillations in the cerebellar cortex of alert mdx mice. These abnormalities were not related to the disrupted expression of calcium-binding proteins in cerebellar PC. We also demonstrate that cerebellar long-term depression is altered in alert mdx mice. Finally, mdx mice displayed a force weakness, mild impairment of motor coordination and balance during behavioural tests. These findings demonstrate the existence of cerebellar dysfunction in mdx mice. A similar cerebellar dysfunction may contribute to the cognitive deficits observed in patients with DMD.
Collapse
Affiliation(s)
- Cynthia Prigogine
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Electrophysiology, Université de Mons, Mons, Belgium
| | | | - Ana Maria Cebolla
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Deconinck
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Brussels, Belgium
| | | | - Philippe Gailly
- Laboratory of Cell Physiology, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Dan
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
- Rehabilitation Hospital Inkendaal, Vlezenbeek, Belgium
| | - Guy Cheron
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Electrophysiology, Université de Mons, Mons, Belgium
| |
Collapse
|
4
|
Kenepp A, Russell-Giller S, Seehra S, Fee R, Hinton VJ. Reading skills over time among children with Duchenne muscular dystrophy. Child Neuropsychol 2024:1-20. [PMID: 39088241 PMCID: PMC11785819 DOI: 10.1080/09297049.2024.2386078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked progressive neuromuscular disorder with a distinct cognitive profile including decreased verbal span. Children with DMD are also at risk for lower scores on academic achievement tests and increased behavioral problems. Longitudinal analyses generally reveal a stable intellectual profile, although attention and behavioral problems may negatively impact longitudinal IQ scores. To date, no study has reported on reading over time in DMD. Reading performance was assessed longitudinally in children with DMD, examining for potential contributions to the trajectory. Retrospective data analysis on assessments completed at baseline, year 2, and year 4 on 26 boys with DMD and 27 unaffected sibling controls (age at baseline: DMD 8 ± 1.4, controls 9 ± 2.6) indicated that children with DMD performed slightly, yet significantly, worse than controls on reading skills, but the longitudinal trajectory of reading skills for children with DMD and controls was not significantly different. Verbal span at time 1 was uniquely associated with later reading skills in children with DMD. Behavior was not associated with declines. The results confirm that children with DMD underperform on reading tasks and align with previous research suggesting that cognitive skills in DMD are stable over time.
Collapse
Affiliation(s)
- Amanda Kenepp
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| | - Shira Russell-Giller
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| | - Sonia Seehra
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| | - Robert Fee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Veronica J Hinton
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| |
Collapse
|
5
|
Peruzzo D, Ciceri T, Mascheretti S, Lampis V, Arrigoni F, Agarwal N, Giubergia A, Villa FM, Crippa A, Nobile M, Mani E, Russo A, D'Angelo MG. Brain Alteration Patterns in Children with Duchenne Muscular Dystrophy: A Machine Learning Approach to Magnetic Resonance Imaging. J Neuromuscul Dis 2024:JND230075. [PMID: 38578898 DOI: 10.3233/jnd-230075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Background Duchenne Muscular Dystrophy (DMD) is a genetic disease in which lack of the dystrophin protein causes progressive muscular weakness, cardiomyopathy and respiratory insufficiency. DMD is often associated with other cognitive and behavioral impairments, however the correlation of abnormal dystrophin expression in the central nervous system with brain structure and functioning remains still unclear. Objective To investigate brain involvement in patients with DMD through a multimodal and multivariate approach accounting for potential comorbidities. Methods We acquired T1-weighted and Diffusion Tensor Imaging data from 18 patients with DMD and 18 age- and sex-matched controls with similar cognitive and behavioral profiles. Cortical thickness, structure volume, fractional anisotropy and mean diffusivity measures were used in a multivariate analysis performed using a Support Vector Machine classifier accounting for potential comorbidities in patients and controls. Results the classification experiment significantly discriminates between the two populations (97.2% accuracy) and the forward model weights showed that DMD mostly affects the microstructural integrity of long fiber bundles, in particular in the cerebellar peduncles (bilaterally), in the posterior thalamic radiation (bilaterally), in the fornix and in the medial lemniscus (bilaterally). We also reported a reduced cortical thickness, mainly in the motor cortex, cingulate cortex, hippocampal area and insula. Conclusions Our study identified a small pattern of alterations in the CNS likely associated with the DMD diagnosis.
Collapse
Affiliation(s)
- Denis Peruzzo
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Tommaso Ciceri
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Sara Mascheretti
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia (PV), Italy
| | - Valentina Lampis
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia (PV), Italy
| | - Filippo Arrigoni
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
- Paediatric Radiology and Neuroradiology Department, V. Buzzi Children's Hospital, Milan, Italy
| | - Nivedita Agarwal
- Diagnostic Imaging and Neuroradiology Unit, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Alice Giubergia
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Filippo Maria Villa
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Alessandro Crippa
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Maria Nobile
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Elisa Mani
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Annamaria Russo
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Maria Grazia D'Angelo
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| |
Collapse
|
6
|
Simon KC, Cadle C, Nakra N, Nagel MC, Malerba P. Age-associated sleep spindle characteristics in Duchenne muscular dystrophy. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae015. [PMID: 38525359 PMCID: PMC10960605 DOI: 10.1093/sleepadvances/zpae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/18/2023] [Indexed: 03/26/2024]
Abstract
Brain oscillations of non-rapid eye movement sleep, including slow oscillations (SO, 0.5-1.5 Hz) and spindles (10-16 Hz), mirror underlying brain maturation across development and are associated with cognition. Hence, age-associated emergence and changes in the electrophysiological properties of these rhythms can lend insight into cortical development, specifically in comparisons between pediatric populations and typically developing peers. We previously evaluated age-associated changes in SOs in male patients with Duchenne muscular dystrophy (DMD), finding a significant age-related decline between 4 and 18 years. While primarily a muscle disorder, male patients with DMD can also have sleep, cognitive, and cortical abnormalities, thought to be driven by altered dystrophin expression in the brain. In this follow-up study, we characterized the age-associated changes in sleep spindles. We found that age-dependent spindle characteristics in patients with DMD, including density, frequency, amplitude, and duration, were consistent with age-associated trends reported in the literature for typically developing controls. Combined with our prior finding of age-associated decline in SOs, our results suggest that SOs, but not spindles, are a candidate intervention target to enhance sleep in patients with DMD.
Collapse
Affiliation(s)
- Katharine C Simon
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, USA
- Pulmonology Department, Children’s Hospital of Orange County, Orange, CA, USA
| | - Chelsea Cadle
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Neal Nakra
- Pulmonology Department, Children’s Hospital of Orange County, Orange, CA, USA
| | - Marni C Nagel
- Department of Pediatric Psychology, Children’s Hospital of Orange County, Orange, CA, USA
| | - Paola Malerba
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, School of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
7
|
Savas-Kalender D, Kurt-Aydin M, Acarol FO, Tarsuslu T, Yis U. Dual task impact on functional mobility and interaction of functional level and balance in patients with Duchenne muscular dystrophy. Gait Posture 2024; 108:282-288. [PMID: 38171184 DOI: 10.1016/j.gaitpost.2023.12.010] [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: 09/07/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Dystrophin, a protein crucial for various brain regions governing higher-order functions like learning and memory is notably absent in individuals with Duchenne muscular dystrophy (DMD). This absence of dystrophin in the brain is believed to underlie cognitive challenges in DMD. Cognitive and motor challenges observed in DMD could potentially hinder the execution of dual tasks. RESEARCH QUESTION Is there a significant correlation between dual-task performance, functional mobility, and balance in children with DMD? METHOD The study included 28 participants (14 DMD, 14 typical development). Timed Up and Go (TUG) test results were recorded for single and dual-task conditions (motor-motor, cognitive-motor). Functional level was assessed using Motor Function Measurement-32 (MFM-32), Brooke Upper Extremity Scale, and Vignos Scale. Balance was evaluated using Balance Master System and Pediatric Functional Reach Test (PFRT). RESULTS Significant differences in TUG test scores across conditions were observed in both DMD and typical development groups (p < 0.05). Children with DMD exhibited longer completion times compared to typical development children (p < 0.05). Among children with DMD, there was a significant correlation between TUG scores in different task conditions and balance assessment (p < 0.05, r = 0.571 to -0.819). Lower MFM-32 scores in DMD children were correlated with worse TUG performance across conditions (p < 0.05, r = 0.586 to -0.868). SIGNIFIANCE This study sheds light on the multifaceted nature of dual-tasking challenges in individuals with DMD, thereby contributing to a deeper understanding of the implications for rehabilitation strategies.
Collapse
Affiliation(s)
| | - Merve Kurt-Aydin
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Izmir Katip Celebi University, Izmir, Turkiye
| | | | - Tulay Tarsuslu
- Faculty of Physical Therapy and Rehabilitation, Dokuz Eylul University, Izmir, Turkiye
| | - Uluc Yis
- Department of Pediatric Neurology, Faculty of Medicine, Dokuz Eylül University, İzmir, Turkiye
| |
Collapse
|
8
|
Weerkamp P, Chieffo D, Collin P, Moriconi F, Papageorgiou A, Vainieri I, Miranda R, Hankinson C, Vogel A, Poncet S, Moss C, Muntoni F, Mercuri E, Hendriksen J. Psychological test usage in duchenne muscular dystrophy: An EU multi-centre study. Eur J Paediatr Neurol 2023; 46:42-47. [PMID: 37423006 DOI: 10.1016/j.ejpn.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023]
Abstract
AIM During the last two decades brain related comorbidities of Duchenne have received growing scientific and clinical interest and therefore systematic assessment of cognition, behaviour and learning is important. This study aims to describe the instruments currently being used in five neuromuscular clinics in Europe as well as the diagnoses being made in these clinics. METHOD A Delphi based procedure was developed by which a questionnaire was sent to the psychologist in five of the seven participating clinics of the Brain Involvement In Dystrophinopathy (BIND) study. Instruments and diagnoses being used were inventoried for three domains of functioning (cognition, behaviour and academics) and three age groups (3-5 years, 6-18 years and adulthood 18+ years). RESULTS Data show wide diversity of tests being used in the five centres at different age groups and different domains. For the intelligence testing there is consensus in using the Wechsler scales, but all other domains such as memory, attention, behavioural problems and reading are tested in very different ways by different instruments in the participating centres. CONCLUSION The heterogeneity of tests and diagnoses being used in current clinical practice underlines the importance for developing a Standard Operating Procedure (SOP) to improve both clinical practice and scientific research over different countries and improve comparative work.
Collapse
Affiliation(s)
- Pien Weerkamp
- Kempenhaeghe Centre for Neurological Learning Disabilities, Heeze, the Netherlands; Maastricht University, School for Mental Health and Neuroscience, Maastricht, the Netherlands
| | - Daniela Chieffo
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Philippe Collin
- Kempenhaeghe Centre for Neurological Learning Disabilities, Heeze, the Netherlands
| | - Federica Moriconi
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | | | - Isabella Vainieri
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Ruben Miranda
- Department of Psychobiology, Universidad Complutense de Madrid, Spain
| | | | - Asmus Vogel
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Sarah Poncet
- Imagine Institute des Maladies Genetiques Necker Enfant Maladies Foundation, Paris, France
| | - Catherine Moss
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Eugenio Mercuri
- Department of Paediatric Neurology, Catholic University, Rome, Italy; Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Jos Hendriksen
- Kempenhaeghe Centre for Neurological Learning Disabilities, Heeze, the Netherlands; Maastricht University, School for Mental Health and Neuroscience, Maastricht, the Netherlands.
| |
Collapse
|
9
|
Wijekoon N, Gonawala L, Ratnayake P, Dissanayaka P, Gunarathne I, Amaratunga D, Liyanage R, Senanayaka S, Wijesekara S, Gunasekara HH, Vanarsa K, Castillo J, Hathout Y, Dalal A, Steinbusch HW, Hoffman E, Mohan C, de Silva KRD. Integrated genomic, proteomic and cognitive assessment in Duchenne Muscular Dystrophy suggest astrocyte centric pathology. Heliyon 2023; 9:e18530. [PMID: 37593636 PMCID: PMC10432191 DOI: 10.1016/j.heliyon.2023.e18530] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023] Open
Abstract
Introduction Documented Duchenne Muscular Dystrophy (DMD) biomarkers are confined to Caucasians and are poor indicators of cognitive difficulties and neuropsychological alterations. Materials and methods This study correlates serum protein signatures with cognitive performance in DMD patients of South Asian origin. Study included 25 DMD patients aged 6-16 years. Cognitive profiles were assessed by Wechsler Intelligence Scale for Children. Serum proteome profiling of 1317 proteins was performed in eight DMD patients and eight age-matched healthy volunteers. Results Among the several novel observations we report, better cognitive performance in DMD was associated with increased serum levels of MMP9 and FN1 but decreased Siglec-3, C4b, and C3b. Worse cognitive performance was associated with increased serum levels of LDH-H1 and PDGF-BB but reduced GDF-11, MMP12, TPSB2, and G1B. Secondly, better cognitive performance in Processing Speed (PSI) and Perceptual Reasoning (PRI) domains was associated with intact Dp116, Dp140, and Dp71 dystrophin isoforms while better performance in Verbal Comprehension (VCI) and Working Memory (WMI) domains was associated with intact Dp116 and Dp140 isoforms. Finally, functional pathways shared with Alzheimer's Disease (AD) point towards an astrocyte-centric model for DMD. Conclusion Astrocytic dysfunction leading to synaptic dysfunction reported previously in AD may be a common pathogenic mechanism underlying both AD and DMD, linking protein alterations to cognitive impairment. This new insight may pave the path towards novel therapeutic approaches targeting reactive astrocytes.
Collapse
Affiliation(s)
- Nalaka Wijekoon
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Lakmal Gonawala
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, The Netherlands
| | | | - Pulasthi Dissanayaka
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
| | - Isuru Gunarathne
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
| | | | - Roshan Liyanage
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
| | | | - Saraji Wijesekara
- Department of Pediatrics, University of Sri Jayewardenepura, 10250, Sri Lanka
- Colombo South Teaching Hospital, 10350, Sri Lanka
| | | | - Kamala Vanarsa
- Department of Bioengineering, University of Houston, Houston, 77204, USA
| | - Jessica Castillo
- Department of Bioengineering, University of Houston, Houston, 77204, USA
| | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, USA
| | - Ashwin Dalal
- Diagnostics Division, Center for DNA Fingerprinting and Diagnostics, India
| | - Harry W.M. Steinbusch
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Eric Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, USA
| | - Chandra Mohan
- Department of Bioengineering, University of Houston, Houston, 77204, USA
| | - K. Ranil D. de Silva
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, The Netherlands
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defence University, Ratmalana, 10390, Sri Lanka
| |
Collapse
|
10
|
Zweyer M, Ohlendieck K, Swandulla D. Verification of Protein Changes Determined by 2D-DIGE Based Proteomics Using Immunofluorescence Microscopy. Methods Mol Biol 2023; 2596:445-464. [PMID: 36378456 DOI: 10.1007/978-1-0716-2831-7_30] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Fluorescence two-dimensional difference gel electrophoresis (2D-DIGE) is a key biochemical method for the comparative analysis of complex protein mixtures. The technique focuses on the identification and characterization of individual protein species following gel electrophoretic separation making it an important analytical tool of top-down proteomics. In order to verify changes in the expression levels of a particular protein, as determined by 2D-DIGE analysis, and evaluate the subcellular localization of the proteoform of interest, immunofluorescence microscopy is very well suited. This chapter describes in detail the preparation of tissue specimens and the process of cryo-sectioning, as well as incubation with primary antibodies and fluorescently labeled secondary antibodies, followed by image analysis. As illustrative examples, the co-detection of immuno-labeled dystrophin and the Y-chromosome in skeletal muscle are shown, and the localization of calbindin in the cerebellum is presented.
Collapse
Affiliation(s)
- Margit Zweyer
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | | |
Collapse
|
11
|
Weerkamp PMM, Mol EM, Sweere DJJ, Schrans DGM, Vermeulen RJ, Klinkenberg S, Hurks PPM, Hendriksen JGM. Wechsler Scale Intelligence Testing in Males with Dystrophinopathies: A Review and Meta-Analysis. Brain Sci 2022; 12:1544. [PMID: 36421868 PMCID: PMC9688319 DOI: 10.3390/brainsci12111544] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Intelligence scores in males with Duchenne Muscular Dystrophy (DMD) and Becker Muscular Dystrophy (BMD) remain a major issue in clinical practice. We performed a literature review and meta-analysis to further delineate the intellectual functioning of dystrophinopathies. METHOD Published, peer-reviewed articles assessing intelligence, using Wechsler Scales, of males with DMD or BMD were searched from 1960 to 2022. Meta-analysis with random-effects models was conducted, assessing weighted, mean effect sizes of full-scale IQ (FSIQ) scores relative to normative data (Mean = 100, Standard Deviation = 15). Post hoc we analysed differences between performance and verbal intelligence scores. RESULTS 43 studies were included, reporting data on 1472 males with dystrophinopathies; with FSIQ scores available for 1234 DMD (k = 32) and 101 BMD (k = 7). DMD males score, on average, one standard deviation below average (FSIQ = 84.76) and significantly lower than BMD (FSIQ = 92.11). Compared to a previous meta-analysis published in 2001, we find, on average, significantly higher FSIQ scores in DMD. CONCLUSION Males with Duchenne have, on average, significantly lower FSIQ scores than BMD males and the general population. Clinicians must consider lower intelligence in dystrophinopathies to ensure good clinical practice.
Collapse
Affiliation(s)
- Pien M. M. Weerkamp
- Expert Centre for Neurological and Developmental Learning Disabilities, Kempenhaeghe, Sterkselseweg 65, 5591 VE Heeze, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, UNS40, 6229 ER Maastricht, The Netherlands
| | - Eva M. Mol
- Expert Centre for Neurological and Developmental Learning Disabilities, Kempenhaeghe, Sterkselseweg 65, 5591 VE Heeze, The Netherlands
- Klimmendaal Academy, Klimmendaal Rehabilitation Specialists, Heijenoordseweg 5, 6813 GG Arnhem, The Netherlands
| | - Dirk J. J. Sweere
- Expert Centre for Neurological and Developmental Learning Disabilities, Kempenhaeghe, Sterkselseweg 65, 5591 VE Heeze, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, UNS40, 6229 ER Maastricht, The Netherlands
| | | | - R. Jeroen Vermeulen
- School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, UNS40, 6229 ER Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Sylvia Klinkenberg
- Expert Centre for Neurological and Developmental Learning Disabilities, Kempenhaeghe, Sterkselseweg 65, 5591 VE Heeze, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, UNS40, 6229 ER Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Petra P. M. Hurks
- School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, UNS40, 6229 ER Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Jos G. M. Hendriksen
- Expert Centre for Neurological and Developmental Learning Disabilities, Kempenhaeghe, Sterkselseweg 65, 5591 VE Heeze, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, UNS40, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
12
|
Barboni MTS, Joachimsthaler A, Roux MJ, Nagy ZZ, Ventura DF, Rendon A, Kremers J, Vaillend C. Retinal dystrophins and the retinopathy of Duchenne muscular dystrophy. Prog Retin Eye Res 2022:101137. [DOI: 10.1016/j.preteyeres.2022.101137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
|
13
|
Language Development in Preschool Duchenne Muscular Dystrophy Boys. Brain Sci 2022; 12:brainsci12091252. [PMID: 36138988 PMCID: PMC9497138 DOI: 10.3390/brainsci12091252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND the present study aims to assess language in preschool-aged Duchenne muscular dystrophy (DMD) boys with normal cognitive quotients, and to establish whether language difficulties are related to attentional aspects or to the involvement of brain dystrophin isoforms. METHODS 20 children aged between 48 and 72 months were assessed with language and attention assessments for preschool children. Nine had a mutation upstream of exon 44, five between 44 and 51, four between 51 and 63, and two after exon 63. A control group comprising 20 age-matched boys with a speech language disorder and normal IQ were also used. RESULTS lexical and syntactic comprehension and denomination were normal in 90% of the boys with Duchenne, while the articulation and repetition of long words, and sentence repetition frequently showed abnormal results (80%). Abnormal results were also found in tests assessing selective and sustained auditory attention. Language difficulties were less frequent in patients with mutations not involving isoforms Dp140 and Dp71. The profile in Duchenne boys was different form the one observed in SLI with no cognitive impairment. CONCLUSION The results of our observational cross-sectional study suggest that early language abilities are frequently abnormal in preschool Duchenne boys and should be assessed regardless of their global neurodevelopmental quotient.
Collapse
|
14
|
Kreko-Pierce T, Pugh JR. Altered Synaptic Transmission and Excitability of Cerebellar Nuclear Neurons in a Mouse Model of Duchenne Muscular Dystrophy. Front Cell Neurosci 2022; 16:926518. [PMID: 35865113 PMCID: PMC9294606 DOI: 10.3389/fncel.2022.926518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is generally regarded as a muscle-wasting disease. However, human patients and animal models of DMD also frequently display non-progressive cognitive deficits and high comorbidity with neurodevelopmental disorders, suggesting impaired central processing. Previous studies have identified the cerebellar circuit, and aberrant inhibitory transmission in Purkinje cells, in particular, as a potential site of dysfunction in the central nervous system (CNS). In this work, we investigate potential dysfunction in the output of the cerebellum, downstream of Purkinje cell (PC) activity. We examined synaptic transmission and firing behavior of excitatory projection neurons of the cerebellar nuclei, the primary output of the cerebellar circuit, in juvenile wild-type and mdx mice, a common mouse model of DMD. Using immunolabeling and electrophysiology, we found a reduced number of PC synaptic contacts, but no change in postsynaptic GABAA receptor expression or clustering in these cells. Furthermore, we found that the replenishment rate of synaptic vesicles in Purkinje terminals is reduced in mdx neurons, suggesting that dysfunction at these synapses may be primarily presynaptic. We also found changes in the excitability of cerebellar nuclear neurons. Specifically, we found greater spontaneous firing but reduced evoked firing from a hyperpolarized baseline in mdx neurons. Analysis of action potential waveforms revealed faster repolarization and greater after-hyperpolarization of evoked action potentials in mdx neurons, suggesting an increased voltage- or calcium- gated potassium current. We did not find evidence of dystrophin protein or messenger RNA (mRNA) expression in wild-type nuclear neurons, suggesting that the changes observed in these cells are likely due to the loss of dystrophin in presynaptic PCs. Together, these data suggest that the loss of dystrophin reduces the dynamic range of synaptic transmission and firing in cerebellar nuclear neurons, potentially disrupting the output of the cerebellar circuit to other brain regions and contributing to cognitive and neurodevelopmental deficits associated with DMD.
Collapse
Affiliation(s)
- Tabita Kreko-Pierce
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Jason R. Pugh
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
15
|
Neuropsychological and behavioral profile in a cohort of Becker Muscular Dystrophy pediatric patients. Neuromuscul Disord 2022; 32:736-742. [DOI: 10.1016/j.nmd.2022.07.402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022]
|
16
|
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: 12] [Impact Index Per Article: 4.0] [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
|
17
|
Passos-Bueno MR, Costa CIS, Zatz M. Dystrophin genetic variants and autism. DISCOVER MENTAL HEALTH 2022; 2:4. [PMID: 37861890 PMCID: PMC10501027 DOI: 10.1007/s44192-022-00008-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/07/2022] [Indexed: 10/21/2023]
Abstract
Loss-of-function variants in the dystrophin gene, a well-known cause of muscular dystrophies, have emerged as a mutational risk mechanism for autism spectrum disorder (ASD), which in turn is a highly prevalent (~ 1%) genetically heterogeneous neurodevelopmental disorder. Although the association of intellectual disability with the dystrophinopathies Duchenne (DMD) and Becker muscular dystrophy (BMD) has been long established, their association with ASD is more recent, and the dystrophin genotype-ASD phenotype correlation is unclear. We therefore present a review of the literature focused on the ASD prevalence among dystrophinopathies, the relevance of the dystrophin isoforms, and most particularly the relevance of the genetic background to the etiology of ASD in these patients. Four families with ASD-DMD/BMD patients are also reported here for the first time. These include a single ASD individual, ASD-discordant and ASD-concordant monozygotic twins, and non-identical ASD triplets. Notably, two unrelated individuals, which were first ascertained because of the ASD phenotype at ages 15 and 5 years respectively, present rare dystrophin variants still poorly characterized, suggesting that some dystrophin variants may compromise the brain more prominently. Whole exome sequencing in these ASD-DMD/BMD individuals together with the literature suggest, although based on preliminary data, a complex and heterogeneous genetic architecture underlying ASD in dystrophinopathies, that include rare variants of large and medium effect. The need for the establishment of a consortia for genomic investigation of ASD-DMD/BMD patients, which may shed light on the genetic architecture of ASD, is discussed.
Collapse
Affiliation(s)
- Maria Rita Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Claudia Ismania Samogy Costa
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Mayana Zatz
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
18
|
Wu WC, Bradley SP, Christie JM, Pugh JR. Mechanisms and Consequences of Cerebellar Purkinje Cell Disinhibition in a Mouse Model of Duchenne Muscular Dystrophy. J Neurosci 2022; 42:2103-2115. [PMID: 35064002 PMCID: PMC8916753 DOI: 10.1523/jneurosci.1256-21.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/13/2021] [Accepted: 01/08/2022] [Indexed: 11/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), the most common form of childhood muscular dystrophy, is caused by mutations in the dystrophin gene. In addition to debilitating muscle degeneration, patients display a range of cognitive deficits thought to result from the loss of dystrophin normally expressed in the brain. While the function of dystrophin in muscle tissue is well characterized, its role in the brain is still poorly understood. The highest expression of dystrophin in the mouse brain is in cerebellar Purkinje cells (PCs), where it colocalizes with GABAA receptor clusters. Using ex vivo electrophysiological recordings from connected molecular layer interneuron (MLI)-PC pairs, we investigated changes in inhibitory synaptic transmission caused by dystrophin deficiency. In male mdx mice (which lack long-form dystrophin), we found that responses at MLI-PC pairs were reduced by ∼60% because of both decreased quantal response amplitude and a reduced number of functional vesicle release sites. Using electron microscopy, we found significantly fewer and smaller anatomically defined inhibitory synapses contacting the soma of PCs in mdx mice, suggesting that dystrophin may play a critical role in synapse formation and/or maintenance. Functionally, we found reduced MLI-evoked pauses in PC firing in acute slices. In vivo recordings from awake mdx mice showed increased sensory-evoked simple spike firing in positively modulating PCs, consistent with reduced feedforward inhibition, but no change in negatively modulating PCs. These data suggest that dystrophin deficiency in PCs disrupts inhibitory signaling in the cerebellar circuit and PC firing patterns, potentially contributing to cognitive and motor deficits observed in mdx mice and DMD patients.SIGNIFICANCE STATEMENT Duchenne muscular dystrophy (DMD) is primarily characterized by progressive muscle weakening caused by genetic mutations in the gene for dystrophin. Dystrophin is also normally expressed in the CNS, and DMD patients experience a range of nonprogressive cognitive deficits. The pathophysiology of CNS neurons resulting from loss of dystrophin and the function of dystrophin in neurons are still poorly understood. Using cerebellar PCs as a model, we found that the loss of dystrophin specifically disrupts the number and strength of inhibitory synaptic connections, suggesting that dystrophin participates in formation and/or maintenance of these synapses. This work provides insight into the function of dystrophin in the CNS and establishes neuronal and synaptic dysfunction, which may underlie cognitive deficits in DMD.
Collapse
Affiliation(s)
- Wan-Chen Wu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Samual P Bradley
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Jason M Christie
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Jason R Pugh
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| |
Collapse
|
19
|
Ohlendieck K, Swandulla D. Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch 2021; 473:1813-1839. [PMID: 34553265 PMCID: PMC8599371 DOI: 10.1007/s00424-021-02623-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Co. Kildare, Maynooth, W23F2H6, Ireland.
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Co. Kildare, Maynooth, W23F2H6, Ireland.
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
20
|
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
|
21
|
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: 15] [Impact Index Per Article: 3.8] [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
|
22
|
Demirci H, Durmus H, Toksoy G, Uslu A, Parman Y, Hanagasi H. Cognition of the mothers of patients with Duchenne muscular dystrophy. Muscle Nerve 2020; 62:710-716. [PMID: 32893363 DOI: 10.1002/mus.27057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/22/2022]
Abstract
Duchenne muscular dystrophy (DMD) has been found to be associated with cognitive impairment. However, few studies have addressed cognitive impairment among mothers of children with DMD. In the present study, the neuropsychological profiles of both carrier mothers (C-Ms) and noncarrier mothers (NC-Ms) were examined, and the findings were compared with healthy control mothers (HC-Ms). There were 90 participants, consisting of 31 C-Ms, 24 NC-Ms, and 35 HC-Ms, each of whom completed a neuropsychological test battery. C-Ms had poorer cognition performance in attention, working memory, immediate verbal memory, visuospatial skills, and executive functions than NC-Ms, and HC-Ms. This study provides evidence that there may be cognitive impairment in mothers of patients with DMD. The cognitive impairment of C-Ms has similarities to that seen in children with DMD.
Collapse
Affiliation(s)
- Hasan Demirci
- Department of Psychiatry, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Hacer Durmus
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Guven Toksoy
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Atilla Uslu
- Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yesim Parman
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hasmet Hanagasi
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
23
|
Persiconi I, Cosmi F, Guadagno NA, Lupo G, De Stefano ME. Dystrophin Is Required for the Proper Timing in Retinal Histogenesis: A Thorough Investigation on the mdx Mouse Model of Duchenne Muscular Dystrophy. Front Neurosci 2020; 14:760. [PMID: 32982660 PMCID: PMC7487415 DOI: 10.3389/fnins.2020.00760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/29/2020] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked muscular disease caused by defective expression of the cytoskeletal protein dystrophin (Dp427). Selected autonomic and central neurons, including retinal neurons, express Dp427 and/or dystrophin shorter isoforms. Because of this, DMD patients may also experience different forms of cognitive impairment, neurological and autonomic disorders, and specific visual defects. DMD-related damages to the nervous system are established during development, suggesting a role for all dystrophin isoforms in neural circuit development and differentiation; however, to date, their function in retinogenesis has never been investigated. In this large-scale study, we analyzed whether the lack of Dp427 affects late retinogenesis in the mdx mouse, the most well studied animal model of DMD. Retinal gene expression and layer maturation, as well as neural cell proliferation, apoptosis, and differentiation, were evaluated in E18 and/or P0, P5, P10, and adult mice. In mdx mice, expression of Capn3, Id3 (E18-P5), and Dtnb (P5) genes, encoding proteins involved in different aspects of retina development and synaptogenesis (e.g., Calpain 3, DNA-binding protein inhibitor-3, and β-dystrobrevin, respectively), was transiently reduced compared to age-matched wild type mice. Concomitantly, a difference in the time required for the retinal ganglion cell layer to reach appropriate thickness was observed (P0–P5). Immunolabeling for specific cell markers also evidenced a significant dysregulation in the number of GABAergic amacrine cells (P5–P10), a transient decrease in the area immunopositive for the Vesicular Glutamate Transporter 1 (VGluT1) during ribbon synapse maturation (P10) and a reduction in the number of calretinin+ retinal ganglion cells (RGCs) (adults). Finally, the number of proliferating retinal progenitor cells (P5–P10) and apoptotic cells (P10) was reduced. These results support the hypothesis of a role for Dp427 during late retinogenesis different from those proposed in consolidated neural circuits. In particular, Dp427 may be involved in shaping specific steps of retina differentiation. Notably, although most of the above described quantitative alterations recover over time, the number of calretinin+ RGCs is reduced only in the mature retina. This suggests that alterations subtler than the timing of retinal maturation may occur, a hypothesis that demands further in-depth functional studies.
Collapse
Affiliation(s)
- Irene Persiconi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Francesca Cosmi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | | | - Giuseppe Lupo
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Maria Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy.,Center for Research in Neurobiology "Daniel Bovet", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
24
|
Xu S, Tang S, Li X, Iyer SR, Lovering RM. Abnormalities in Brain and Muscle Microstructure and Neurochemistry of the DMD Rat Measured by in vivo Diffusion Tensor Imaging and High Resolution Localized 1H MRS. Front Neurosci 2020; 14:739. [PMID: 32760246 PMCID: PMC7372970 DOI: 10.3389/fnins.2020.00739] [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: 03/18/2020] [Accepted: 06/22/2020] [Indexed: 12/03/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disorder caused by the lack of dystrophin with progressive degeneration of skeletal muscles. Most studies regarding DMD understandably focus on muscle, but dystrophin is also expressed in the central nervous system, potentially resulting in cognitive and behavioral changes. Animal models are being used for developing more comprehensive neuromonitoring protocols and clinical image acquisition procedures. The recently developed DMD rat is an animal model that parallels the progressive muscle wasting seen in DMD. Here, we studied the brain and temporalis muscle structure and neurochemistry of wild type (WT) and dystrophic (DMD) rats using magnetic resonance imaging and spectroscopy. Both structural and neurochemistry alterations were observed in the DMD rat brain and the temporalis muscle. There was a decrease in absolute brain volume (WT = 1579 mm3; DMD = 1501 mm3; p = 0.039, Cohen’s d = 1.867), but not normalized (WT = 4.27; DMD = 4.02; p = 0.306) brain volume. Diffusion tensor imaging (DTI) revealed structural alterations in the DMD temporalis muscle, with increased mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD). In the DMD rat thalamus, DTI revealed an increase in fractional anisotropy (FA) and a decrease in RD. Smaller normalized brain volume correlated to severity of muscle dystrophy (r = −0.975). Neurochemical changes in the DMD rat brain included increased GABA and NAA in the prefrontal cortex, and GABA in the hippocampus. Such findings could indicate disturbed motor and sensory signaling, resulting in a dysfunctional GABAergic neurotransmission, and an unstable osmoregulation in the dystrophin-null brain.
Collapse
Affiliation(s)
- Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States.,Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shiyu Tang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States.,Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Xin Li
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
25
|
Peristeri E, Aloizou AM, Keramida P, Tsouris Z, Siokas V, Mentis AFA, Dardiotis E. Cognitive Deficits in Myopathies. Int J Mol Sci 2020; 21:ijms21113795. [PMID: 32471196 PMCID: PMC7312055 DOI: 10.3390/ijms21113795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
Myopathies represent a wide spectrum of heterogeneous diseases mainly characterized by the abnormal structure or functioning of skeletal muscle. The current paper provides a comprehensive overview of cognitive deficits observed in various myopathies by consulting the main libraries (Pubmed, Scopus and Google Scholar). This review focuses on the causal classification of myopathies and concomitant cognitive deficits. In most studies, cognitive deficits have been found after clinical observations while lesions were also present in brain imaging. Most studies refer to hereditary myopathies, mainly Duchenne muscular dystrophy (DMD), and myotonic dystrophies (MDs); therefore, most of the overview will focus on these subtypes of myopathies. Most recent bibliographical sources have been preferred.
Collapse
Affiliation(s)
- Eleni Peristeri
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, PC 41110 Larissa, Greece; (E.P.); (A.-M.A.); (P.K.); (Z.T.); (V.S.)
| | - Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, PC 41110 Larissa, Greece; (E.P.); (A.-M.A.); (P.K.); (Z.T.); (V.S.)
| | - Paraskevi Keramida
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, PC 41110 Larissa, Greece; (E.P.); (A.-M.A.); (P.K.); (Z.T.); (V.S.)
| | - Zisis Tsouris
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, PC 41110 Larissa, Greece; (E.P.); (A.-M.A.); (P.K.); (Z.T.); (V.S.)
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, PC 41110 Larissa, Greece; (E.P.); (A.-M.A.); (P.K.); (Z.T.); (V.S.)
| | - Alexios-Fotios A. Mentis
- Public Health Laboratories, Hellenic Pasteur Institute, PC 11521 Athens, Greece;
- Department of Microbiology, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, PC 41110 Larissa, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, PC 41110 Larissa, Greece; (E.P.); (A.-M.A.); (P.K.); (Z.T.); (V.S.)
- Correspondence: ; Tel.:+ 30-241-350-1137
| |
Collapse
|
26
|
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: 50] [Impact Index Per Article: 10.0] [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
|
27
|
Hippocampal synaptic and membrane function in the DBA/2J-mdx mouse model of Duchenne muscular dystrophy. Mol Cell Neurosci 2020; 104:103482. [PMID: 32171922 DOI: 10.1016/j.mcn.2020.103482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Dystrophin deficiency is associated with alterations in cell physiology. The functional consequences of dystrophin deficiency are particularly severe for muscle physiology, as observed in Duchenne muscle dystrophy (DMD). DMD is caused by the absence of a 427 kDa isoform of dystrophin. However, in addition to muscular dystrophy symptoms, DMD is frequently associated with memory and attention deficits and epilepsy. While this may be associated with a role for dystrophin in neuronal physiology, it is not clear what neuronal alterations are linked with DMD. Our work shows that CA1 pyramidal neurons from DBA/2J-mdx mice have increased afterhyperpolarization compared to WT controls. All the other electrotonic and electrogenic membrane properties were unaffected by this genotype. Finally, basal synaptic transmission, short-term and long-term synaptic plasticity at Schaffer collateral to CA1 glutamatergic synapses were unchanged between mdx and WT controls. These data show that the excitatory component of hippocampal activity is largely preserved in DBA/2J-mdx mice. Further studies, extending the investigation to the inhibitory GABAergic function, may provide a more complete picture of the functional, network alterations underlying impaired cognition in DMD. In addition, the investigation of changes in neuronal single conductance biophysical properties associated with this genotype, is required to identify the functional alterations associated with dystrophin deficiency and clarify its role in neuronal function.
Collapse
|
28
|
Cognitive impairment appears progressive in the mdx mouse. Neuromuscul Disord 2020; 30:368-388. [PMID: 32360405 PMCID: PMC7306157 DOI: 10.1016/j.nmd.2020.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 11/22/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive muscle wasting disease caused by mutations in the DMD gene, which encodes the large cytoskeletal protein dystrophin. Approximately one-third of DMD patient's exhibit cognitive problems yet it is unknown if cognitive impairments worsen with age. The mdx mouse model is deficient in dystrophin demonstrates cognitive abnormalities, but no studies have investigated this longitudinally. We assessed the consequences of dystrophin deficiency on brain morphology and cognition in male mdx mice. We utilised non-invasive methods to monitor CNS pathology with an aim to identify changes longitudinally (between 4 and 18 months old) which could be used as outcome measures. MRI identified a total brain volume (TBV) increase in control mice with ageing (p < 0.05); but the mdx mice TBV increased significantly more (p < 0.01). Voxel-based morphometry (VBM) identified decreases in grey matter volume, particularly in the hippocampus of the mdx brain, most noticeable from 12 months onwards, as were enlarged lateral ventricles in mdx mice. The caudate putamen of older mdx mice showed increases in T2- relaxometry which may be considered as evidence of increased water content. Hippocampal spatial learning and memory was decreased in mdx mice, particularly long-term memory, which progressively worsened with age. The novel object recognition (NOR) task highlighted elevated anxiety-related behaviour in older mdx mice. Our studies suggest that dystrophin deficiency causes a progressive cognitive impairment in mice (compared to ageing control mice), becoming evident at late disease stages, and may explain why progressive CNS symptoms are not obvious in DMD patients.
Collapse
|
29
|
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: 85] [Impact Index Per Article: 17.0] [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
|
30
|
Stay TL, Miterko LN, Arancillo M, Lin T, Sillitoe RV. In vivo cerebellar circuit function is disrupted in an mdx mouse model of Duchenne muscular dystrophy. Dis Model Mech 2019; 13:dmm040840. [PMID: 31704708 PMCID: PMC6906634 DOI: 10.1242/dmm.040840] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a debilitating and ultimately lethal disease involving progressive muscle degeneration and neurological dysfunction. DMD is caused by mutations in the dystrophin gene, which result in extremely low or total loss of dystrophin protein expression. In the brain, dystrophin is heavily localized to cerebellar Purkinje cells, which control motor and non-motor functions. In vitro experiments in mouse Purkinje cells revealed that loss of dystrophin leads to low firing rates and high spiking variability. However, it is still unclear how the loss of dystrophin affects cerebellar function in the intact brain. Here, we used in vivo electrophysiology to record Purkinje cells and cerebellar nuclear neurons in awake and anesthetized female mdx (also known as Dmd) mice. Purkinje cell simple spike firing rate is significantly lower in mdx mice compared to controls. Although simple spike firing regularity is not affected, complex spike regularity is increased in mdx mutants. Mean firing rate in cerebellar nuclear neurons is not altered in mdx mice, but their local firing pattern is irregular. Based on the relatively well-preserved cytoarchitecture in the mdx cerebellum, our data suggest that faulty signals across the circuit between Purkinje cells and cerebellar nuclei drive the abnormal firing activity. The in vivo requirements of dystrophin during cerebellar circuit communication could help explain the motor and cognitive anomalies seen in individuals with DMD.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Trace L Stay
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Lauren N Miterko
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Marife Arancillo
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Tao Lin
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
31
|
Dickson PE, Mittleman G. Visual Discrimination, Serial Reversal, and Extinction Learning in the mdx Mouse. Front Behav Neurosci 2019; 13:200. [PMID: 31543764 PMCID: PMC6728792 DOI: 10.3389/fnbeh.2019.00200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy and the most common neuromuscular disorder. In addition to neuromuscular consequences, some individuals with DMD experience global intellectual dysfunction and executive dysfunction of unknown mechanistic origin. The cognitive profile of the mdx mouse, the most commonly used mouse model of DMD, has been incompletely characterized and has never been assessed using the touchscreen operant conditioning paradigm. The touchscreen paradigm allows the use of protocols that are virtually identical to those used in human cognitive testing and may, therefore, provide the most translational paradigm for quantifying mouse cognitive function. In the present study, we used the touchscreen paradigm to assess the effects of the mdx mutation on visual discrimination learning, serial reversal learning, and extinction learning. To enable measuring task-dependent learning and memory processes while holding demands on sensory-driven information processing constant, we developed equally salient visual stimuli and used them on all experimental stages. Acquisition of the initial pairwise visual discrimination was facilitated in mdx mice relative to wildtype littermates; this effect was not explained by genotypic differences in impulsivity, motivation, or motor deficits. The mdx mutation had no effect on serial reversal or extinction learning. Together, findings from this study and previous studies suggest that mdx effects on cognitive function are task-specific and may be influenced by discrimination type (spatial, visual), reward type (food, escape from a non-preferred environment), sex, and genetic background.
Collapse
Affiliation(s)
| | - Guy Mittleman
- Department of Psychological Science, Ball State University, Muncie, IN, United States
| |
Collapse
|
32
|
Sato K, Tachikawa M, Watanabe M, Miyauchi E, Uchida Y, Terasaki T. Identification of Blood-Brain Barrier-Permeable Proteins Derived from a Peripheral Organ: In Vivo and in Vitro Evidence of Blood-to-Brain Transport of Creatine Kinase. Mol Pharm 2019; 16:247-257. [PMID: 30495961 DOI: 10.1021/acs.molpharmaceut.8b00975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Certain proteins, such as inflammatory cytokines, that are released from injured or diseased organs are transported from the circulating blood through the blood-brain barrier (BBB) into the brain and contribute to the pathogenesis of related central nervous system dysfunctions. However, little is known about the protein transport mechanisms involved in the central nervous system dysfunctions. The aims of the present study were to identify BBB-permeable protein(s) derived from liver and to clarify their transport characteristics at the BBB. After administration of biotin-labeled liver cytosolic protein fraction to mice in vivo, we identified 9 biotin-labeled proteins in the brain. Among them, we focused here on creatine kinase (CK). In vitro uptake studies with human brain microvessel endothelial cells (hCMEC/D3 cells) showed preferential uptake of muscle-type CK (CK-MM) compared with brain-type CK (CK-BB) at the BBB. Integration plot analysis revealed that CK-MM readily penetrated into brain parenchyma from the circulating blood across the BBB. The uptake of CK-MM by hCMEC/D3 cells was decreased at 4 °C and in the presence of clathrin- and caveolin-dependent endocytosis inhibitors. These results indicate that entry of CK into the brain is mediated by a transport system(s) at the BBB.
Collapse
Affiliation(s)
- Kazuki Sato
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Michitoshi Watanabe
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Eisuke Miyauchi
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| |
Collapse
|
33
|
Helleringer R, Le Verger D, Li X, Izabelle C, Chaussenot R, Belmaati-Cherkaoui M, Dammak R, Decottignies P, Daniel H, Galante M, Vaillend C. Cerebellar synapse properties and cerebellum-dependent motor and non-motor performance in Dp71-null mice. Dis Model Mech 2018; 11:dmm.033258. [PMID: 29895670 PMCID: PMC6078407 DOI: 10.1242/dmm.033258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 06/04/2018] [Indexed: 02/04/2023] Open
Abstract
Recent emphasis has been placed on the role that cerebellar dysfunctions could have in the genesis of cognitive deficits in Duchenne muscular dystrophy (DMD). However, relevant genotype-phenotype analyses are missing to define whether cerebellar defects underlie the severe cases of intellectual deficiency that have been associated with genetic loss of the smallest product of the dmd gene, the Dp71 dystrophin. To determine for the first time whether Dp71 loss could affect cerebellar physiology and functions, we have used patch-clamp electrophysiological recordings in acute cerebellar slices and a cerebellum-dependent behavioral test battery addressing cerebellum-dependent motor and non-motor functions in Dp71-null transgenic mice. We found that Dp71 deficiency selectively enhances excitatory transmission at glutamatergic synapses formed by climbing fibers (CFs) on Purkinje neurons, but not at those formed by parallel fibers. Altered basal neurotransmission at CFs was associated with impairments in synaptic plasticity and clustering of the scaffolding postsynaptic density protein PSD-95. At the behavioral level, Dp71-null mice showed some improvements in motor coordination and were unimpaired for muscle force, static and dynamic equilibrium, motivation in high-motor demand and synchronization learning. Dp71-null mice displayed altered strategies in goal-oriented navigation tasks, however, suggesting a deficit in the cerebellum-dependent processing of the procedural components of spatial learning, which could contribute to the visuospatial deficits identified in this model. In all, the observed deficits suggest that Dp71 loss alters cerebellar synapse function and cerebellum-dependent navigation strategies without being detrimental for motor functions. Summary: Dp71 is the most prominent dystrophin gene product in the adult brain. Here, multiple approaches including behavioral tests and electrophysiology are adopted to explore the role of Dp71 in the cerebellum.
Collapse
Affiliation(s)
- Romain Helleringer
- Molecules and Circuits Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Delphine Le Verger
- Cognition and Behavior Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Xia Li
- Molecules and Circuits Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Charlotte Izabelle
- Cognition and Behavior Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Rémi Chaussenot
- Cognition and Behavior Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Mehdi Belmaati-Cherkaoui
- Cognition and Behavior Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Raoudha Dammak
- Molecules and Circuits Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Paulette Decottignies
- Molecules and Circuits Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Hervé Daniel
- Molecules and Circuits Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Micaela Galante
- Molecules and Circuits Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| | - Cyrille Vaillend
- Cognition and Behavior Department, Paris-Saclay Institute of Neuroscience (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, 91405 Orsay, France
| |
Collapse
|
34
|
Cognitive Event-Related Potentials (P300) and Cognitive Impairment in Duchenne Muscular Dystrophy. NEUROPHYSIOLOGY+ 2018. [DOI: 10.1007/s11062-018-9695-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
35
|
Doorenweerd N, Hooijmans M, Schubert SA, Webb AG, Straathof CSM, van Zwet EW, van Buchem MA, Verschuuren JJGM, Hendriksen JGM, Niks EH, Kan HE. Proton Magnetic Resonance Spectroscopy Indicates Preserved Cerebral Biochemical Composition in Duchenne Muscular Dystrophy Patients. J Neuromuscul Dis 2018; 4:53-58. [PMID: 28269793 DOI: 10.3233/jnd-160201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is caused by the absence of dystrophin. DMD is associated with specific learning and behavioural disabilities. In the brain, dystrophin is associated with GABAA receptors and aquaporin-4 in neurons and astrocytes, respectively, but little is known about its function. OBJECTIVE AND METHODS In this study we aimed to compare the biochemical composition between patients and healthy controls in brain regions that are naturally rich in dystrophin using magnetic resonance spectroscopy. Given previous conflicting results obtained at clinical field strengths, we obtained data using a 7 Tesla system with associated higher signal-to-noise ratio and spectral resolution. RESULTS Results indicated unchanged biochemical composition in all regions investigated, and increased variance in glutamate in the frontal cortex.
Collapse
Affiliation(s)
- Nathalie Doorenweerd
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden, The Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa Hooijmans
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephanie A Schubert
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew G Webb
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Chiara S M Straathof
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik W van Zwet
- Department of Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mark A van Buchem
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jos G M Hendriksen
- Department of Neurological Learning Disabilities, Kempenhaeghe Epilepsy Center, Heeze, The Netherlands.,Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermien E Kan
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| |
Collapse
|
36
|
Vicari S, Piccini G, Mercuri E, Battini R, Chieffo D, Bulgheroni S, Pecini C, Lucibello S, Lenzi S, Moriconi F, Pane M, D’Amico A, Astrea G, Baranello G, Riva D, Cioni G, Alfieri P. Implicit learning deficit in children with Duchenne muscular dystrophy: Evidence for a cerebellar cognitive impairment? PLoS One 2018; 13:e0191164. [PMID: 29338029 PMCID: PMC5770044 DOI: 10.1371/journal.pone.0191164] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 12/31/2017] [Indexed: 11/19/2022] Open
Abstract
This study aimed at comparing implicit sequence learning in individuals affected by Duchenne Muscular Dystrophy without intellectual disability and age-matched typically developing children. A modified version of the Serial Reaction Time task was administered to 32 Duchenne children and 37 controls of comparable chronological age. The Duchenne group showed a reduced rate of implicit learning even if in the absence of global intellectual disability. This finding provides further evidence of the involvement of specific aspects of cognitive function in Duchenne muscular dystrophy and on its possible neurobiological substrate.
Collapse
Affiliation(s)
- Stefano Vicari
- Department of Neuroscience, Child Neuropsychiatric Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giorgia Piccini
- Department of Neuroscience, Child Neuropsychiatric Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Catholic University and Nemo Center, Rome, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Daniela Chieffo
- Pediatric Neurology Unit, Catholic University and Nemo Center, Rome, Italy
| | - Sara Bulgheroni
- Developmental Neurology Division, IRCCS Fondazione Istituto Neurologico C. Besta, Milan, Italy
| | - Chiara Pecini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
| | - Simona Lucibello
- Pediatric Neurology Unit, Catholic University and Nemo Center, Rome, Italy
| | - Sara Lenzi
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
| | - Federica Moriconi
- Pediatric Neurology Unit, Catholic University and Nemo Center, Rome, Italy
| | - Marika Pane
- Pediatric Neurology Unit, Catholic University and Nemo Center, Rome, Italy
| | - Adele D’Amico
- Department of Neurosciences, Neuromuscular and Neurodegenerative Diseases Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Guja Astrea
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
| | - Giovanni Baranello
- Developmental Neurology Division, IRCCS Fondazione Istituto Neurologico C. Besta, Milan, Italy
| | - Daria Riva
- Developmental Neurology Division, IRCCS Fondazione Istituto Neurologico C. Besta, Milan, Italy
| | - Giovanni Cioni
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Paolo Alfieri
- Department of Neuroscience, Child Neuropsychiatric Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- * E-mail:
| |
Collapse
|
37
|
Abstract
Duchenne muscular dystrophy is a highly progressive neuromuscular disorder caused by primary abnormalities in the Dmd gene encoding the membrane cytoskeletal protein dystrophin. Dystrophinopathies are multi-systems disorders that are characterized by severe skeletal muscle wasting, with loss of independent ambulation in the early teenage years, followed by cardio-respiratory complications and premature death. Nonprogressive cognitive impairments are estimated to affect approximately one-third of dystrophic children. To identify the molecular mechanisms behind the impaired brain function in dystrophinopathy, liquid chromatography-based mass spectrometry offers an unbiased and technology-driven approach. In this chapter, we give a detailed description of a label-free mass spectrometric method to investigate proteome-wide changes in the dystrophin-deficient brain from a genetic mouse model of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Sandra Murphy
- Department of Biology, Maynooth University, National University of Ireland Maynooth, Callan Building, North Campus, Maynooth, Co. Kildare, Ireland
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland Maynooth, Callan Building, North Campus, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
38
|
Battini R, Chieffo D, Bulgheroni S, Piccini G, Pecini C, Lucibello S, Lenzi S, Moriconi F, Pane M, Astrea G, Baranello G, Alfieri P, Vicari S, Riva D, Cioni G, Mercuri E. Cognitive profile in Duchenne muscular dystrophy boys without intellectual disability: The role of executive functions. Neuromuscul Disord 2017; 28:122-128. [PMID: 29305139 DOI: 10.1016/j.nmd.2017.11.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/19/2017] [Accepted: 11/29/2017] [Indexed: 11/16/2022]
Abstract
The aim of our prospective observational study was to assess profiles of cognitive function and a possible impairment of executive functions in a cohort of boys with Duchenne muscular dystrophy without intellectual and behavior disability. Forty Duchenne boys (range of age: 6 years to 11 years and 6 months) were assessed by Wechsler Intelligence scale and battery of tests including tasks assessing working memory and executive functions (inhibition and switching, problem solving and planning). In our cohort some aspects of cognitive function were often impaired. These included multitasking, problem solving, inhibition and working memory necessary to plan and direct goal oriented behavior. Our results support the suggestion that aspects of cognitive function could be impaired even in boys without intellectual disability and support the hypothesis that executive functions may play an important role in specific aspects of cognitive impairment in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- R Battini
- Pediatric Neurology Unit, Fondazione Policlinico Gemelli, and Nemo Center, UCSC Rome, Italy; Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
| | - D Chieffo
- Pediatric Neurology Unit, Fondazione Policlinico Gemelli, and Nemo Center, UCSC Rome, Italy
| | - S Bulgheroni
- Developmental Neurology Division, IRCCS Fondazione Istituto Neurologico C. Besta, Milan, Italy
| | - G Piccini
- Unit of Child Neuropsichiatry, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - C Pecini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
| | - S Lucibello
- Pediatric Neurology Unit, Fondazione Policlinico Gemelli, and Nemo Center, UCSC Rome, Italy
| | - S Lenzi
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
| | - F Moriconi
- Pediatric Neurology Unit, Fondazione Policlinico Gemelli, and Nemo Center, UCSC Rome, Italy
| | - M Pane
- Pediatric Neurology Unit, Fondazione Policlinico Gemelli, and Nemo Center, UCSC Rome, Italy
| | - G Astrea
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy
| | - G Baranello
- Developmental Neurology Division, IRCCS Fondazione Istituto Neurologico C. Besta, Milan, Italy
| | - P Alfieri
- Unit of Child Neuropsichiatry, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - S Vicari
- Unit of Child Neuropsichiatry, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - D Riva
- Developmental Neurology Division, IRCCS Fondazione Istituto Neurologico C. Besta, Milan, Italy
| | - G Cioni
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone (Pisa), Italy; Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - E Mercuri
- Pediatric Neurology Unit, Fondazione Policlinico Gemelli, and Nemo Center, UCSC Rome, Italy.
| |
Collapse
|
39
|
Mavrogeni S, Pons R, Nikas I, Papadopoulos G, Verganelakis DA, Kolovou G, Chrousos GP. Brain and heart magnetic resonance imaging/spectroscopy in duchenne muscular dystrophy. Eur J Clin Invest 2017; 47. [PMID: 28981141 DOI: 10.1111/eci.12842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 10/02/2017] [Indexed: 01/04/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked muscle disorder characterized by progressive and irreversible loss of muscular function. As muscular disease progresses, the repair mechanisms cannot compensate for cellular damage, leading inevitably to necrosis and progressive replacement by fibrous and fatty tissue. Cardiomyopathy and respiratory failure are the main causes of death in DMD. In addition to the well-described muscle and heart disease, cognitive dysfunction affects around 30% of DMD boys. Myocardial fibrosis, assessed by late gadolinium enhancement (LGE), using cardiovascular magnetic resonance imaging (CMR), is an early marker of heart involvement in both DMD patients and female carriers. In parallel, brain MRI identifies smaller total brain volume, smaller grey matter volume, lower white matter fractional anisotropy and higher white matter radial diffusivity in DMD patients. The in vivo brain evaluation of mdx mice, a surrogate animal model of DMD, showed an increased inorganic phosphate (P(i))/phosphocreatine (PCr) and pH. In this paper, we propose a holistic approach using techniques of magnetic resonance imaging, spectroscopy and diffusion tensor imaging as a tool to create a "heart and brain imaging map" in DMD patients that could potentially facilitate the patients' risk stratification and also future research studies in the field.
Collapse
Affiliation(s)
| | - Roser Pons
- 1st Department of Pediatrics, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | - Ioannis Nikas
- 1st Department of Pediatrics, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | - George Papadopoulos
- 1st Department of Pediatrics, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | - Dimitrios A Verganelakis
- 1st Department of Pediatrics, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | | | - George P Chrousos
- 1st Department of Pediatrics, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| |
Collapse
|
40
|
Timing and localization of human dystrophin isoform expression provide insights into the cognitive phenotype of Duchenne muscular dystrophy. Sci Rep 2017; 7:12575. [PMID: 28974727 PMCID: PMC5626779 DOI: 10.1038/s41598-017-12981-5] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 09/13/2017] [Indexed: 01/14/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a muscular dystrophy with high incidence of learning and behavioural problems and is associated with neurodevelopmental disorders. To gain more insights into the role of dystrophin in this cognitive phenotype, we performed a comprehensive analysis of the expression patterns of dystrophin isoforms across human brain development, using unique transcriptomic data from Allen Human Brain and BrainSpan atlases. Dystrophin isoforms show large changes in expression through life with pronounced differences between the foetal and adult human brain. The Dp140 isoform was expressed in the cerebral cortex only in foetal life stages, while in the cerebellum it was also expressed postnatally. The Purkinje isoform Dp427p was virtually absent. The expression of dystrophin isoforms was significantly associated with genes implicated in neurodevelopmental disorders, like autism spectrum disorders or attention-deficit hyper-activity disorders, which are known to be associated to DMD. We also identified relevant functional associations of the different isoforms, like an association with axon guidance or neuron differentiation during early development. Our results point to the crucial role of several dystrophin isoforms in the development and function of the human brain.
Collapse
|
41
|
Wang JZ, Wu P, Shi ZM, Xu YL, Liu ZJ. The AAV-mediated and RNA-guided CRISPR/Cas9 system for gene therapy of DMD and BMD. Brain Dev 2017; 39:547-556. [PMID: 28390761 DOI: 10.1016/j.braindev.2017.03.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/13/2017] [Accepted: 03/19/2017] [Indexed: 12/26/2022]
Abstract
Mutations in the dystrophin gene (Dmd) result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD), which afflict many newborn boys. In 2016, Brain and Development published several interesting articles on DMD treatment with antisense oligonucleotide, kinase inhibitor, and prednisolone. Even more strikingly, three articles in the issue 6271 of Science in 2016 provide new insights into gene therapy of DMD and BMD via the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9). In brief, adeno-associated virus (AAV) vectors transport guided RNAs (gRNAs) and Cas9 into mdx mouse model, gRNAs recognize the mutated Dmd exon 23 (having a stop codon), and Cas9 cut the mutated exon 23 off the Dmd gene. These manipulations restored expression of truncated but partially functional dystrophin, improved skeletal and cardiac muscle function, and increased survival of mdx mice significantly. This review concisely summarized the related advancements and discussed their primary implications in the future gene therapy of DMD, including AAV-vector selection, gRNA designing, Cas9 optimization, dystrophin-restoration efficiency, administration routes, and systemic and long-term therapeutic efficacy. Future orientations, including off-target effects, safety concerns, immune responses, precision medicine, and Dmd-editing in the brain (potentially blocked by the blood-brain barrier) were also elucidated briefly. Collectively, the AAV-mediated and RNA-guided CRISPR/Cas9 system has major superiorities compared with traditional gene therapy, and might contribute to the treatment of DMD and BMD substantially in the near future.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China.
| | - Peng Wu
- Department of Social Science, Hebei University of Engineering, Handan 056038, PR China
| | - Zhi-Min Shi
- College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| | - Yan-Li Xu
- College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| | - Zhi-Jun Liu
- College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China.
| |
Collapse
|
42
|
Lombardi L, Persiconi I, Gallo A, Hoogenraad CC, De Stefano ME. NGF-dependent axon growth and regeneration are altered in sympathetic neurons of dystrophic mdx mice. Mol Cell Neurosci 2017; 80:1-17. [PMID: 28161362 DOI: 10.1016/j.mcn.2017.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 12/09/2016] [Accepted: 01/29/2017] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal disease, determined by lack of dystrophin (Dp427), a muscular cytoskeletal protein also expressed by selected neuronal populations. Consequently, besides muscular wasting, both human patients and DMD animal models suffer several neural disorders. In previous studies on the superior cervical ganglion (SCG) of wild type and dystrophic mdx mice (Lombardi et al. 2008), we hypothesized that Dp427 could play some role in NGF-dependent axonal growth, both during development and adulthood. To address this issue, we first analyzed axon regeneration potentials of SCG neurons of both genotypes after axotomy in vivo. While noradrenergic innervation of mdx mouse submandibular gland, main source of nerve growth factor (NGF), recovered similarly to wild type, iris innervation (muscular target) never did. We, therefore, evaluated whether dystrophic SCG neurons were poorly responsive to NGF, especially at low concentration. Following in vitro axotomy in the presence of either 10 or 50ng/ml NGF, the number of regenerated axons in mdx mouse neuron cultures was indeed reduced, compared to wild type, at the lower concentration. Neurite growth parameters (i.e. number, length), growth cone dynamics and NGF/TrkA receptor signaling in differentiating neurons (not injured) were also significantly reduced when cultured with 10ng/ml NGF, but also with higher NGF concentrations. In conclusion, we propose a role for Dp427 in NGF-dependent cytoskeletal dynamics associated to growth cone advancement, possibly through indirect stabilization of TrkA receptors. Considering NGF activity in nervous system development/remodeling, this aspect could concur in some of the described DMD-associated neural dysfunctions.
Collapse
Affiliation(s)
- Loredana Lombardi
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy
| | - Irene Persiconi
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy
| | - Alessandra Gallo
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, 3584CH Utrecht, The Netherlands
| | - Maria Egle De Stefano
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy; Center for Research in Neurobiology "Daniel Bovet", Sapienza Università di Roma, 00185 Roma, Italy.
| |
Collapse
|
43
|
Filippini M, Guerra A, Negosanti A, Santi S, Sarajlija J, Musti MA, Gobbi G, Lassonde M, Pini A. Mismatch Negativity Recording in Children With Duchenne Muscular Dystrophy: A Preliminary Study Integrating Neurophysiological and Neuropsychological Results. J Child Neurol 2016; 31:1468-1474. [PMID: 27422795 DOI: 10.1177/0883073816656404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 05/26/2016] [Indexed: 11/15/2022]
Abstract
Many studies on Duchenne muscular dystrophy children support the hypothesis of a specific neuropsychological phenotype affecting mostly phonological skills. This prospective study aimed to shed light on the role of phonological abilities. Fourteen Duchenne muscular dystrophy children and 7 healthy children underwent mismatch negativity. Moreover, verbal intelligence, visuospatial attention, immediate verbal memory, working memory, grammar, vocabulary, visuomotor skills, reading, text comprehension, writing, and arithmetic were tested in Duchenne muscular dystrophy children. No significant difference between control and Duchenne muscular dystrophy children was found neither for mismatch negativity amplitude (P = .191 and .116, respectively) nor for latency (P = .135). Eight (57.14%) patients showed an impairment of immediate verbal memory and of visuomotor skills, 7 (63.64%) patients had a deficit in writing and arithmetic skills, even with a mean normal intelligence quotient. Taken together, the results put in evidence a heterogeneous neuropsychological profile not explainable on the basis of a phonological deficit.
Collapse
Affiliation(s)
- Melissa Filippini
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | - Angelo Guerra
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | | | - Sara Santi
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | - Jasenka Sarajlija
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | | | - Giuseppe Gobbi
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | - Maryse Lassonde
- Department of Psychology, Université de Montréal, Montréal, Canada
| | - Antonella Pini
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| |
Collapse
|
44
|
Digit Span Performance in Children with Dystrophinopathy: A Verbal Span or Working Memory Contribution? J Int Neuropsychol Soc 2016; 22:777-84. [PMID: 27268852 DOI: 10.1017/s1355617716000461] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVES In a large cohort of boys with dystrophinopathies and their unaffected siblings, we examined whether consistently observed performance on digit span is due primarily to a verbal span or executive deficit. We additionally assessed whether digit span performance contributed to the observed variability in reading performance noted in this population. METHODS Performance of 170 boys with dystrophinopathy was compared to 95 unaffected sibling controls on measures of verbal function, reading, and digit span. Maximum digit span forward (DSF) and backward (DSB) lengths were converted to Z-scores using normative data. Independent sample t tests, analysis of variance, and hierarchical multiple regression were run (α=0.05). RESULTS Probands performed worse than controls on digit span, even after accounting for differences in general verbal function (p<.0001). Differences were significant for both DSF (p<.005) and DSB (p<.0001) span length, and an interaction effect yielded significantly worse DSB compared with DSF (p=.01). Reading performance was also lower in probands (p<.0001). The contribution of general level of verbal function, and forward and backward span lengths, did not vary between groups. CONCLUSIONS In boys with dystrophinopathy, decreased performance on digit span appears to be due to both decreased span forward (measuring verbal span only) and backward (measuring verbal span and working memory). The extent to which sibling controls exhibited better performance compared to the probands was significantly greater for backward span when compared with forward span. Thus, immediate verbal memory and executive control are differentially compromised among boys with dystrophinopathy, and both of these abilities independently contribute to reading performance. (JINS, 2016, 22, 777-784).
Collapse
|
45
|
Rae MG, O'Malley D. Cognitive dysfunction in Duchenne muscular dystrophy: a possible role for neuromodulatory immune molecules. J Neurophysiol 2016; 116:1304-15. [PMID: 27385793 DOI: 10.1152/jn.00248.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/29/2016] [Indexed: 11/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X chromosome-linked disease characterized by progressive physical disability, immobility, and premature death in affected boys. Underlying the devastating symptoms of DMD is the loss of dystrophin, a structural protein that connects the extracellular matrix to the cell cytoskeleton and provides protection against contraction-induced damage in muscle cells, leading to chronic peripheral inflammation. However, dystrophin is also expressed in neurons within specific brain regions, including the hippocampus, a structure associated with learning and memory formation. Linked to this, a subset of boys with DMD exhibit nonprogressing cognitive dysfunction, with deficits in verbal, short-term, and working memory. Furthermore, in the genetically comparable dystrophin-deficient mdx mouse model of DMD, some, but not all, types of learning and memory are deficient, and specific deficits in synaptogenesis and channel clustering at synapses has been noted. Little consideration has been devoted to the cognitive deficits associated with DMD compared with the research conducted into the peripheral effects of dystrophin deficiency. Therefore, this review focuses on what is known about the role of full-length dystrophin (Dp427) in hippocampal neurons. The importance of dystrophin in learning and memory is assessed, and the potential importance that inflammatory mediators, which are chronically elevated in dystrophinopathies, may have on hippocampal function is also evaluated.
Collapse
Affiliation(s)
- Mark G Rae
- Department of Physiology, University College Cork, Cork, Ireland; and
| | - Dervla O'Malley
- Department of Physiology, University College Cork, Cork, Ireland; and APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
46
|
Russo FB, Cugola FR, Fernandes IR, Pignatari GC, Beltrão-Braga PCB. Induced pluripotent stem cells for modeling neurological disorders. World J Transplant 2015; 5:209-221. [PMID: 26722648 PMCID: PMC4689931 DOI: 10.5500/wjt.v5.i4.209] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/23/2015] [Accepted: 09/28/2015] [Indexed: 02/05/2023] Open
Abstract
Several diseases have been successfully modeled since the development of induced pluripotent stem cell (iPSC) technology in 2006. Since then, methods for increased reprogramming efficiency and cell culture maintenance have been optimized and many protocols for differentiating stem cell lines have been successfully developed, allowing the generation of several cellular subtypes in vitro. Gene editing technologies have also greatly advanced lately, enhancing disease-specific phenotypes by creating isogenic cell lines, allowing mutations to be corrected in affected samples or inserted in control lines. Neurological disorders have benefited the most from iPSC-disease modeling for its capability for generating disease-relevant cell types in vitro from the central nervous system, such as neurons and glial cells, otherwise only available from post-mortem samples. Patient-specific iPSC-derived neural cells can recapitulate the phenotypes of these diseases and therefore, considerably enrich our understanding of pathogenesis, disease mechanism and facilitate the development of drug screening platforms for novel therapeutic targets. Here, we review the accomplishments and the current progress in human neurological disorders by using iPSC modeling for Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, spinal muscular atrophy, amyotrophic lateral sclerosis, duchenne muscular dystrophy, schizophrenia and autism spectrum disorders, which include Timothy syndrome, Fragile X syndrome, Angelman syndrome, Prader-Willi syndrome, Phelan-McDermid, Rett syndrome as well as Nonsyndromic Autism.
Collapse
|
47
|
Miranda R, Nagapin F, Bozon B, Laroche S, Aubin T, Vaillend C. Altered social behavior and ultrasonic communication in the dystrophin-deficient mdx mouse model of Duchenne muscular dystrophy. Mol Autism 2015; 6:60. [PMID: 26527530 PMCID: PMC4627616 DOI: 10.1186/s13229-015-0053-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 10/22/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The Duchenne and Becker muscular dystrophies (DMD, BMD) show significant comorbid diagnosis for autism, and the genomic sequences encoding the proteins responsible for these diseases, the dystrophin and associated proteins, have been proposed as new candidate risk loci for autism. Dystrophin is expressed not only in muscles but also in central inhibitory synapses in the cerebellum, hippocampus, amygdala, and cerebral cortex, where it contributes to the organization of autism-associated trans-synaptic neurexin-neuroligin complexes and to the clustering of synaptic gamma-aminobutyric acid (GABA)A receptors. While brain defects due to dystrophin loss are associated with deficits in cognitive and executive functions, communication skills and social behavior, only a subpopulation of DMD patients meet the criteria for autism, suggesting that mutations in the dystrophin gene may confer a vulnerability to autism. The loss of dystrophin in the mdx mouse model of DMD has been associated with cognitive and emotional alterations, but social behavior and communication abilities have never been studied in this model. METHODS Here, we carried out the first in-depth analysis of social behavior and ultrasonic communication in dystrophin-deficient mdx mice, using a range of socially relevant paradigms involving various degrees of executive and cognitive demands, from simple presentation of sexual olfactory stimuli to social choice situations and direct encounters with female and male mice of various genotypes. RESULTS We identified context-specific alterations in social behavior and ultrasonic vocal communication in mdx mice during direct encounters in novel environments. Social behavior disturbances depended on intruders' genotype and behavior, suggesting alterations in executive functions and adaptive behaviors, and were associated with selective alterations of the development, rate, acoustic properties, and use of the ultrasonic vocal repertoire. CONCLUSIONS This first evidence that a mutation impeding expression of brain dystrophin affects social behavior and communication sheds new light on critical cognitive, emotional, and conative factors contributing to the development of autistic-like traits in this disease model.
Collapse
Affiliation(s)
- Rubén Miranda
- CNRS, Neuroscience Paris Saclay Institute, UMR 9197, Orsay, 91405 France ; Univ Paris-Sud, UMR 9197, Orsay, 91405 France ; Present address: Department of Psychobiology, Universidad Complutense de Madrid, Ciudad Universitaria, 28040 Madrid, Spain
| | - Flora Nagapin
- CNRS, Neuroscience Paris Saclay Institute, UMR 9197, Orsay, 91405 France ; Univ Paris-Sud, UMR 9197, Orsay, 91405 France
| | - Bruno Bozon
- CNRS, Neuroscience Paris Saclay Institute, UMR 9197, Orsay, 91405 France ; Univ Paris-Sud, UMR 9197, Orsay, 91405 France
| | - Serge Laroche
- CNRS, Neuroscience Paris Saclay Institute, UMR 9197, Orsay, 91405 France ; Univ Paris-Sud, UMR 9197, Orsay, 91405 France
| | - Thierry Aubin
- CNRS, Neuroscience Paris Saclay Institute, UMR 9197, Orsay, 91405 France ; Univ Paris-Sud, UMR 9197, Orsay, 91405 France
| | - Cyrille Vaillend
- CNRS, Neuroscience Paris Saclay Institute, UMR 9197, Orsay, 91405 France ; Univ Paris-Sud, UMR 9197, Orsay, 91405 France
| |
Collapse
|
48
|
Chieffo D, Brogna C, Berardinelli A, D'Angelo G, Mallardi M, D'Amico A, Alfieri P, Mercuri E, Pane M. Early Neurodevelopmental Findings Predict School Age Cognitive Abilities in Duchenne Muscular Dystrophy: A Longitudinal Study. PLoS One 2015; 10:e0133214. [PMID: 26275215 PMCID: PMC4537199 DOI: 10.1371/journal.pone.0133214] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 06/23/2015] [Indexed: 12/28/2022] Open
Abstract
Objective Neurodevelopmental and cognitive difficulties are known to occur frequently in boys with Duchenne muscular dystrophy but so far none of the published studies have reported both early neurodevelopmental assessments and cognitive tests in the same cohort. The aim of the present longitudinal study was to establish the correlation between early neurodevelopmental assessments performed in preschool boys and the cognitive scales performed at school age or later. Methods We performed cognitive tests at school age (mean age 5.7 year ±1.7 SD) (69 months+19 SD) in a cohort of Duchenne boys, previously assessed using the Griffiths scales before the age of 4 years (mean age when the Griffiths scales were performed 30 months ±8.9 SD). Results The range of total Developmental quotients on the Griffiths ranged between 56 and 116 (mean 89 ± 15.6 SD). The total Intelligence Quotients on the Wechsler scales ranged between 35 and 119 (mean 87 ± 17.2 SD). There was a significant correlation between the findings on the two scales. P = <0.0001. When we subdivided the cohort according to site of mutations, there was a difference between boys with mutations upstream exon 44 and those with mutations in exon 44–45 affecting Dp140 on both Developmental and Intelligence Quotient (p 0.01 and p 0,003 respectively). Conclusions Our results confirm that Duchenne boys tend to slightly underperform on both neurodevelopmental and cognitive assessments. Early neurodevelopmental findings correlated with the cognitive results obtained at school age with a clear concordance between subscales exploring similar domains on the two scales.
Collapse
Affiliation(s)
- Daniela Chieffo
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Claudia Brogna
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | | | | | - Maria Mallardi
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Adele D'Amico
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, Rome, Italy
| | - Paolo Alfieri
- Child and Adolescence Neuropsychiatry Unit, Department of Neuroscience Bambino Gesù Children's Hospital, Rome, Italy
| | - Eugenio Mercuri
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Marika Pane
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| |
Collapse
|
49
|
Nichols B, Takeda S, Yokota T. Nonmechanical Roles of Dystrophin and Associated Proteins in Exercise, Neuromuscular Junctions, and Brains. Brain Sci 2015; 5:275-98. [PMID: 26230713 PMCID: PMC4588140 DOI: 10.3390/brainsci5030275] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/29/2015] [Accepted: 07/21/2015] [Indexed: 02/06/2023] Open
Abstract
Dystrophin-glycoprotein complex (DGC) is an important structural unit in skeletal muscle that connects the cytoskeleton (f-actin) of a muscle fiber to the extracellular matrix (ECM). Several muscular dystrophies, such as Duchenne muscular dystrophy, Becker muscular dystrophy, congenital muscular dystrophies (dystroglycanopathies), and limb-girdle muscular dystrophies (sarcoglycanopathies), are caused by mutations in the different DGC components. Although many early studies indicated DGC plays a crucial mechanical role in maintaining the structural integrity of skeletal muscle, recent studies identified novel roles of DGC. Beyond a mechanical role, these DGC members play important signaling roles and act as a scaffold for various signaling pathways. For example, neuronal nitric oxide synthase (nNOS), which is localized at the muscle membrane by DGC members (dystrophin and syntrophins), plays an important role in the regulation of the blood flow during exercise. DGC also plays important roles at the neuromuscular junction (NMJ) and in the brain. In this review, we will focus on recently identified roles of DGC particularly in exercise and the brain.
Collapse
Affiliation(s)
- Bailey Nichols
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry.
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan.
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry.
- Muscular Dystrophy Canada Research Chair, 8812-112 St, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
50
|
Chaussenot R, Edeline JM, Le Bec B, El Massioui N, Laroche S, Vaillend C. Cognitive dysfunction in the dystrophin-deficient mouse model of Duchenne muscular dystrophy: A reappraisal from sensory to executive processes. Neurobiol Learn Mem 2015; 124:111-22. [PMID: 26190833 DOI: 10.1016/j.nlm.2015.07.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 07/09/2015] [Accepted: 07/10/2015] [Indexed: 01/08/2023]
Abstract
Duchenne muscular dystrophy (DMD) is associated with language disabilities and deficits in learning and memory, leading to intellectual disability in a patient subpopulation. Recent studies suggest the presence of broader deficits affecting information processing, short-term memory and executive functions. While the absence of the full-length dystrophin (Dp427) is a common feature in all patients, variable mutation profiles may additionally alter distinct dystrophin-gene products encoded by separate promoters. However, the nature of the cognitive dysfunctions specifically associated with the loss of distinct brain dystrophins is unclear. Here we show that the loss of the full-length brain dystrophin in mdx mice does not modify the perception and sensorimotor gating of auditory inputs, as assessed using auditory brainstem recordings and prepulse inhibition of startle reflex. In contrast, both acquisition and long-term retention of cued and trace fear memories were impaired in mdx mice, suggesting alteration in a functional circuit including the amygdala. Spatial learning in the water maze revealed reduced path efficiency, suggesting qualitative alteration in mdx mice learning strategy. However, spatial working memory performance and cognitive flexibility challenged in various behavioral paradigms in water and radial-arm mazes were unimpaired. The full-length brain dystrophin therefore appears to play a role during acquisition of associative learning as well as in general processes involved in memory consolidation, but no overt involvement in working memory and/or executive functions could be demonstrated in spatial learning tasks.
Collapse
Affiliation(s)
- Rémi Chaussenot
- Paris-Saclay Neuroscience Institute, UMR 9197, CNRS, F-91405 Orsay, France; Univ. Paris-Sud, UMR 9197, F-91405 Orsay, France; Université Paris-Saclay, France
| | - Jean-Marc Edeline
- Paris-Saclay Neuroscience Institute, UMR 9197, CNRS, F-91405 Orsay, France; Univ. Paris-Sud, UMR 9197, F-91405 Orsay, France; Université Paris-Saclay, France
| | - Benoit Le Bec
- Paris-Saclay Neuroscience Institute, UMR 9197, CNRS, F-91405 Orsay, France; Univ. Paris-Sud, UMR 9197, F-91405 Orsay, France; Université Paris-Saclay, France
| | - Nicole El Massioui
- Paris-Saclay Neuroscience Institute, UMR 9197, CNRS, F-91405 Orsay, France; Univ. Paris-Sud, UMR 9197, F-91405 Orsay, France; Université Paris-Saclay, France
| | - Serge Laroche
- Paris-Saclay Neuroscience Institute, UMR 9197, CNRS, F-91405 Orsay, France; Univ. Paris-Sud, UMR 9197, F-91405 Orsay, France; Université Paris-Saclay, France
| | - Cyrille Vaillend
- Paris-Saclay Neuroscience Institute, UMR 9197, CNRS, F-91405 Orsay, France; Univ. Paris-Sud, UMR 9197, F-91405 Orsay, France; Université Paris-Saclay, France.
| |
Collapse
|