1
|
Ryan-Phillips F, Henehan L, Ramdas S, Palace J, Beeson D, Dong YY. Assessing the Utility of ColabFold and AlphaMissense in Determining Missense Variant Pathogenicity for Congenital Myasthenic Syndromes. Biomedicines 2024; 12:2549. [PMID: 39595115 PMCID: PMC11592069 DOI: 10.3390/biomedicines12112549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Congenital myasthenic syndromes (CMSs) are caused by variants in >30 genes with increasing numbers of variants of unknown significance (VUS) discovered by next-generation sequencing. Establishing VUS pathogenicity requires in vitro studies that slow diagnosis and treatment initiation. The recently developed protein structure prediction software AlphaFold2/ColabFold has revolutionized structural biology; such predictions have also been leveraged in AlphaMissense, which predicts ClinVar variant pathogenicity with 90% accuracy. Few reports, however, have tested these tools on rigorously characterized clinical data. We therefore assessed ColabFold and AlphaMissense as diagnostic aids for CMSs, using variants of the CHRN genes that encode the nicotinic acetylcholine receptor (nAChR). METHODS Utilizing a dataset of 61 clinically validated CHRN variants, (1) we evaluated the possibility of a ColabFold metric (either predicted structural disruption, prediction confidence, or prediction quality) that distinguishes variant pathogenicity; (2) we assessed AlphaMissense's ability to differentiate variant pathogenicity; and (3) we compared AlphaMissense to the existing pathogenicity prediction programs AlamutVP and EVE. RESULTS Analyzing the variant effects on ColabFold CHRN structure prediction, prediction confidence, and prediction quality did not yield any reliable pathogenicity indicative metric. However, AlphaMissense predicted variant pathogenicity with 63.93% accuracy in our dataset-a much greater proportion than AlamutVP (27.87%) and EVE (28.33%). CONCLUSIONS Emerging in silico tools can revolutionize genetic disease diagnosis-however, improvement, refinement, and clinical validation are imperative prior to practical acquisition.
Collapse
Affiliation(s)
- Finlay Ryan-Phillips
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
| | - Leighann Henehan
- Neurology Department, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Sithara Ramdas
- Department of Paediatric Neurology, John Radcliffe Hospital, Oxford OX3 9DU, UK;
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
- Neurology Department, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
| | - Yin Yao Dong
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
2
|
Smeets N, Gheldof A, Dequeker B, Poleur M, Maldonado Slootjes S, Van Parijs V, Deconinck N, Dontaine P, Alonso-Jimenez A, De Bleecker J, De Ridder W, Herdewyn S, Paquay S, Vanlander A, De Waele L, Peirens G, Beysen D, Claeys KG, Dubuisson N, Hansen I, Remiche G, Seneca S, Bissay V, Régal L. Congenital Myasthenic Syndromes in Belgium: Genetic and Clinical Characterization of Pediatric and Adult Patients. Pediatr Neurol 2024; 158:57-65. [PMID: 38964204 DOI: 10.1016/j.pediatrneurol.2024.06.002] [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: 03/10/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Congenital myasthenic syndromes (CMS) are a group of genetic disorders characterized by impaired neuromuscular transmission. CMS typically present at a young age with fatigable muscle weakness, often with an abnormal response after repetitive nerve stimulation (RNS). Pharmacologic treatment can improve symptoms, depending on the underlying defect. Prevalence is likely underestimated. This study reports on patients with CMS followed in Belgium in 2022. METHODS Data were gathered retrospectively from the medical charts. Only likely pathogenic and pathogenic variants were included in the analysis. RESULTS We identified 37 patients, resulting in an estimated prevalence of 3.19 per 1,000,000. The patients harbored pathogenic variants in CHRNE, RAPSN, DOK7, PREPL, CHRNB1, CHRNG, COLQ, MUSK, CHRND, GFPT1, and GMPPB. CHRNE was the most commonly affected gene. Most patients showed disease onset at birth, during infancy, or during childhood. Symptom onset was at adult age in seven patients, caused by variants in CHRNE, DOK7, MUSK, CHRND, and GMPPB. Severity and distribution of weakness varied, as did the presence of respiratory involvement, feeding problems, and extraneuromuscular manifestations. RNS was performed in 23 patients of whom 18 demonstrated a pathologic decrement. Most treatment responses were predictable based on the genotype. CONCLUSIONS This is the first pooled characterization of patients with CMS in Belgium. We broaden the phenotypical spectrum of pathogenic variants in CHRNE with adult-onset CMS. Systematically documenting larger cohorts of patients with CMS can aid in better clinical characterization and earlier recognition of this rare disease. We emphasize the importance of establishing a molecular genetic diagnosis to tailor treatment choices.
Collapse
Affiliation(s)
- Nathalie Smeets
- Child Neurology Unit, Department of Pediatrics, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium.
| | - Alexander Gheldof
- Center of Medical Genetics, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Bart Dequeker
- Center of Medical Genetics, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Margaux Poleur
- University Department of Neurology, Citadelle Hospital of Liège, Liège, Belgium
| | | | - Vinciane Van Parijs
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Nicolas Deconinck
- Department of Pediatric Neurology, Queen Fabiola Children's University Hospital, Université Libre De Bruxelles, Brussels, Belgium
| | - Pauline Dontaine
- Department of Pediatric Neurology, Queen Fabiola Children's University Hospital, Université Libre De Bruxelles, Brussels, Belgium
| | - Alicia Alonso-Jimenez
- Department of Neurology, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Jan De Bleecker
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Willem De Ridder
- Department of Neurology, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Sarah Herdewyn
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Stéphanie Paquay
- Department of Neuropediatrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arnaud Vanlander
- Department of Pediatric Neurology and Metabolic Diseases, Ghent University Hospital, Ghent, Belgium
| | - Liesbeth De Waele
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Department of Pediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Geertrui Peirens
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Department of Pediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Diane Beysen
- Department of Pediatric Neurology, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium; Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Nicolas Dubuisson
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Isabelle Hansen
- Department of Neurology, University of Liège, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - Gauthier Remiche
- Department of Neurology, Hôpital Universitaire de Bruxelles - Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Sara Seneca
- Center of Medical Genetics, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Véronique Bissay
- NEUR Research Group and Department of Neurology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Luc Régal
- Child Neurology Unit, Department of Pediatrics, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
3
|
Natera-de Benito D, Pugliese A, Polavarapu K, Guergueltcheva V, Tournev I, Todorova A, Afonso Ribeiro J, Fernández-Mayoralas DM, Ortez C, Martorell L, Estévez-Arias B, Matalonga L, Laurie S, Jou C, Lau J, Thompson R, Shen X, Engel AG, Nascimento A, Lochmüller H, Selcen D. Advancing the Understanding of Vesicle-Associated Membrane Protein 1-Related Congenital Myasthenic Syndrome: Phenotypic Insights, Favorable Response to 3,4-Diaminopyridine, and Clinical Characterization of Five New Cases. Pediatr Neurol 2024; 157:5-13. [PMID: 38833907 PMCID: PMC11257830 DOI: 10.1016/j.pediatrneurol.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/27/2024] [Accepted: 04/28/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Congenital myasthenic syndromes (CMS) are a group of inherited neuromuscular junction (NMJ) disorders arising from gene variants encoding diverse NMJ proteins. Recently, the VAMP1 gene, responsible for encoding the vesicle-associated membrane protein 1 (VAMP1), has been associated with CMS. METHODS This study presents a characterization of five new individuals with VAMP1-related CMS, providing insights into the phenotype. RESULTS The individuals with VAMP1-related CMS exhibited early disease onset, presenting symptoms prenatally or during the neonatal period, alongside severe respiratory involvement and feeding difficulties. Generalized weakness at birth was a common feature, and none of the individuals achieved independent walking ability. Notably, all cases exhibited scoliosis. The clinical course remained stable, without typical exacerbations seen in other CMS types. The response to anticholinesterase inhibitors and salbutamol was only partial, but the addition of 3,4-diaminopyridine (3,4-DAP) led to significant and substantial improvements, suggesting therapeutic benefits of 3,4-DAP for managing VAMP1-related CMS symptoms. Noteworthy is the identification of the VAMP1 (NM_014231.5): c.340delA; p.Ile114SerfsTer72 as a founder variant in the Iberian Peninsula and Latin America. CONCLUSIONS This study contributes valuable insights into VAMP1-related CMS, emphasizing their early onset, arthrogryposis, facial and generalized weakness, respiratory involvement, and feeding difficulties. Furthermore, the potential efficacy of 3,4-DAP as a useful therapeutic option warrants further exploration. The findings have implications for clinical management and genetic counseling in affected individuals. Additional research is necessary to elucidate the long-term outcomes of VAMP1-related CMS.
Collapse
Affiliation(s)
- Daniel Natera-de Benito
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain; Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.
| | - Alessia Pugliese
- IRCCS Centro Neurolesi "Bonino-Pulejo", Neurology Unit, Messina, Italy
| | - Kiran Polavarapu
- Children' s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Velina Guergueltcheva
- Clinic of Neurology, University Hospital Sofiamed, Sofia University St. Kliment Ohridski, Sofia, Bulgaria
| | - Ivailo Tournev
- Department of Neurology, University Hospital "Alexandrovska", Medical University, Sofia, Bulgaria; Department of Cognitive Science and Psychology, New Bulgarian University, Sofia, Bulgaria
| | - Albena Todorova
- Genetic Medico-Diagnostic Laboratory "Genica", Sofia, Bulgaria; Department of Medical Chemistry and Biochemistry, Sofia Medical University, Sofia, Bulgaria
| | | | | | - Carlos Ortez
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain; Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Loreto Martorell
- Department of Genetic and Molecular Medicine-IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Berta Estévez-Arias
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain; Laboratory of Neurogenetics and Molecular Medicine-IPER, Sant Joan de Deu Research Institute, Barcelona, Spain
| | | | - Steven Laurie
- Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - Cristina Jou
- Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain; Department of Pathology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jarred Lau
- Children' s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Rachel Thompson
- Children' s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Xinming Shen
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Andrew G Engel
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Andres Nascimento
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu, Barcelona, Spain; Applied Research in Neuromuscular Diseases, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Hanns Lochmüller
- Children' s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada; Laboratory of Neurogenetics and Molecular Medicine-IPER, Sant Joan de Deu Research Institute, Barcelona, Spain; Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada; Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Duygu Selcen
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
4
|
Li H, Teng J, Hibbs RE. Structural switch in acetylcholine receptors in developing muscle. Nature 2024; 632:1174-1180. [PMID: 39085615 DOI: 10.1038/s41586-024-07774-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
During development, motor neurons originating in the brainstem and spinal cord form elaborate synapses with skeletal muscle fibres1. These neurons release acetylcholine (ACh), which binds to nicotinic ACh receptors (AChRs) on the muscle, initiating contraction. Two types of AChR are present in developing muscle cells, and their differential expression serves as a hallmark of neuromuscular synapse maturation2-4. The structural principles underlying the switch from fetal to adult muscle receptors are unknown. Here, we present high-resolution structures of both fetal and adult muscle nicotinic AChRs, isolated from bovine skeletal muscle in developmental transition. These structures, obtained in the absence and presence of ACh, provide a structural context for understanding how fetal versus adult receptor isoforms are tuned for synapse development versus the all-or-none signalling required for high-fidelity skeletal muscle contraction. We find that ACh affinity differences are driven by binding site access, channel conductance is tuned by widespread surface electrostatics and open duration changes result from intrasubunit interactions and structural flexibility. The structures further reveal pathogenic mechanisms underlying congenital myasthenic syndromes.
Collapse
Affiliation(s)
- Huanhuan Li
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Jinfeng Teng
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Ryan E Hibbs
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
5
|
Tian S, Sun H, Gao FF, Zhang K, Nan J, Niu M, Jia X, Xu G, Ge W. Genetic analysis of a family affected by congenital myasthenic syndrome due to a Novel mutation in the SLC5A7 gene. BMC Neurol 2024; 24:206. [PMID: 38886633 PMCID: PMC11181541 DOI: 10.1186/s12883-024-03716-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Mutations in the SLC5A7 gene cause congenital myasthenia, a rare genetic disorder. Mutation points in the SLC5A7 gene differ among individuals and encompass various genetic variations; however, exon deletion variants have yet to be reported in related cases. This study aims to explore the clinical phenotype and genetic traits of a patient with congenital myasthenic syndrome due to SLC5A7 gene variation and those of their family members. CASE PRESENTATION We describe a case of a Chinese male with congenital myasthenic syndrome presenting fluctuating limb weakness. Genetic testing revealed a heterozygous deletion mutation spanning exons 1-9 in the SLC5A7 gene. QPCR confirmed a deletion in exon 9 of the SLC5A7 gene in the patient's mother and brother. Clinical symptoms of myasthenia improved following treatment with pyridostigmine. CONCLUSION Exons 1, 5, and 9 of the SLC5A7 gene encode the choline transporter's transmembrane region. Mutations in these exons can impact the stability and plasma membrane levels of the choline transporter. Thus, a heterozygous deletion in exons 1-9 of the SLC5A7 gene could be the pathogenic cause for this patient. In patients exhibiting fluctuating weakness, positive RNS, and seronegativity for myasthenia gravis antibodies, a detailed family history should be considered, and enhanced genetic testing is recommended to determine the cause.
Collapse
Affiliation(s)
- Sheng Tian
- Xuzhou Medical University, Xuzhou, China
| | - Huan Sun
- Xuzhou Medical University, Xuzhou, China
| | | | - Kang Zhang
- Xuzhou Medical University, Xuzhou, China
| | - Jing Nan
- Xuzhou Medical University, Xuzhou, China
| | - Mu Niu
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical College Affiliated Hospital, Xuzhou, China
| | - Xiao Jia
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical College Affiliated Hospital, Xuzhou, China
| | - Gang Xu
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical College Affiliated Hospital, Xuzhou, China
| | - Wei Ge
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical College Affiliated Hospital, Xuzhou, China.
| |
Collapse
|
6
|
Henehan L, Beeson D, Palace J. Congenital myasthenic syndromes. Pract Neurol 2024; 24:185-187. [PMID: 38631903 DOI: 10.1136/pn-2024-004098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2024] [Indexed: 04/19/2024]
Affiliation(s)
- Leighann Henehan
- Department of Clinical Neurology, John Radcliffe Hospital, Oxford, Oxfordshire, UK
| | - David Beeson
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jacqueline Palace
- Department of Clinical Neurology, John Radcliffe Hospital, Oxford, Oxfordshire, UK
| |
Collapse
|
7
|
Bharambe N, Li Z, Seiferth D, Balakrishna AM, Biggin PC, Basak S. Cryo-EM structures of prokaryotic ligand-gated ion channel GLIC provide insights into gating in a lipid environment. Nat Commun 2024; 15:2967. [PMID: 38580666 PMCID: PMC10997623 DOI: 10.1038/s41467-024-47370-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
GLIC, a proton-activated prokaryotic ligand-gated ion channel, served as a model system for understanding the eukaryotic counterparts due to their structural and functional similarities. Despite extensive studies conducted on GLIC, the molecular mechanism of channel gating in the lipid environment requires further investigation. Here, we present the cryo-EM structures of nanodisc-reconstituted GLIC at neutral and acidic pH in the resolution range of 2.6 - 3.4 Å. In our apo state at pH 7.5, the extracellular domain (ECD) displays conformational variations compared to the existing apo structures. At pH 4.0, three distinct conformational states (C1, C2 and O states) are identified. The protonated structures exhibit a compacted and counter-clockwise rotated ECD compared with our apo state. A gradual widening of the pore in the TMD is observed upon reducing the pH, with the widest pore in O state, accompanied by several layers of water pentagons. The pore radius and molecular dynamics (MD) simulations suggest that the O state represents an open conductive state. We also observe state-dependent interactions between several lipids and proteins that may be involved in the regulation of channel gating. Our results provide comprehensive insights into the importance of lipids impact on gating.
Collapse
Affiliation(s)
- Nikhil Bharambe
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Zhuowen Li
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - David Seiferth
- Structural Bioinformatics and Computational Biochemistry, Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - Philip C Biggin
- Structural Bioinformatics and Computational Biochemistry, Department of Biochemistry, University of Oxford, Oxford, UK
| | - Sandip Basak
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
- NTU Institute of Structural Biology, Nanyang Technological University, Singapore, 639798, Singapore.
| |
Collapse
|
8
|
Núñez-Carpintero I, Rigau M, Bosio M, O'Connor E, Spendiff S, Azuma Y, Topf A, Thompson R, 't Hoen PAC, Chamova T, Tournev I, Guergueltcheva V, Laurie S, Beltran S, Capella-Gutiérrez S, Cirillo D, Lochmüller H, Valencia A. Rare disease research workflow using multilayer networks elucidates the molecular determinants of severity in Congenital Myasthenic Syndromes. Nat Commun 2024; 15:1227. [PMID: 38418480 PMCID: PMC10902324 DOI: 10.1038/s41467-024-45099-0] [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: 12/21/2022] [Accepted: 01/15/2024] [Indexed: 03/01/2024] Open
Abstract
Exploring the molecular basis of disease severity in rare disease scenarios is a challenging task provided the limitations on data availability. Causative genes have been described for Congenital Myasthenic Syndromes (CMS), a group of diverse minority neuromuscular junction (NMJ) disorders; yet a molecular explanation for the phenotypic severity differences remains unclear. Here, we present a workflow to explore the functional relationships between CMS causal genes and altered genes from each patient, based on multilayer network community detection analysis of complementary biomedical information provided by relevant data sources, namely protein-protein interactions, pathways and metabolomics. Our results show that CMS severity can be ascribed to the personalized impairment of extracellular matrix components and postsynaptic modulators of acetylcholine receptor (AChR) clustering. This work showcases how coupling multilayer network analysis with personalized -omics information provides molecular explanations to the varying severity of rare diseases; paving the way for sorting out similar cases in other rare diseases.
Collapse
Affiliation(s)
- Iker Núñez-Carpintero
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain
| | - Maria Rigau
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Mattia Bosio
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain
- Coordination Unit Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Barcelona Supercomputing Center, Barcelona, Spain
| | - Emily O'Connor
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Sally Spendiff
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Yoshiteru Azuma
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Ana Topf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Rachel Thompson
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Peter A C 't Hoen
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Teodora Chamova
- Department of Neurology, Expert Centre for Hereditary Neurologic and Metabolic Disorders, Alexandrovska University Hospital, Medical University-Sofia, Sofia, Bulgaria
| | - Ivailo Tournev
- Department of Neurology, Expert Centre for Hereditary Neurologic and Metabolic Disorders, Alexandrovska University Hospital, Medical University-Sofia, Sofia, Bulgaria
- Department of Cognitive Science and Psychology, New Bulgarian University, Sofia, 1618, Bulgaria
| | - Velina Guergueltcheva
- Clinic of Neurology, University Hospital Sofiamed, Sofia University St. Kliment Ohridski, Sofia, Bulgaria
| | - Steven Laurie
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Sergi Beltran
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Salvador Capella-Gutiérrez
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain
- Coordination Unit Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Barcelona Supercomputing Center, Barcelona, Spain
| | - Davide Cirillo
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain.
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain
- Coordination Unit Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Barcelona Supercomputing Center, Barcelona, Spain
- ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain
| |
Collapse
|
9
|
Cossins J, Kozma I, Canzonetta C, Hawkins A, Beeson D, Sepulveda P, Dong Y. Dose escalation pre-clinical trial of novel DOK7-AAV in mouse model of DOK7 congenital myasthenia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579626. [PMID: 38405691 PMCID: PMC10888934 DOI: 10.1101/2024.02.09.579626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Congenital myasthenic syndromes (CMS) are a group of inherited disorders characterised by defective neuromuscular transmission and fatigable muscle weakness. Mutations in DOK7 , a gene encoding a post-synaptic protein crucial in the formation and stabilisation of the neuromuscular junction (NMJ), rank among the leading three prevalent causes of CMS in diverse populations globally. The majority of DOK7 CMS patients experience varying degrees of disability despite receiving optimised treatment, necessitating the development of improved therapeutic approaches. Here we executed a dose escalation pre-clinical trial using a DOK7-CMS mouse model to assess the efficacy of Amp-101, an innovative AAV gene replacement therapy. Amp-101 is based on AAVrh74 and contains human DOK7 cDNA under the control of a muscle-restricted promoter. We show that at doses 6x10 13 vg/kg and 1x10 14 vg/kg, Amp-101 generated enlarged NMJs and rescued the very severe phenotype of the model. Treated mice became at least as strong as WT littermates and the diaphragm and tibialis anterior muscles displayed robust expression of DOK7. This data suggests that Amp-101 is a promising candidate to move forward to clinic trials.
Collapse
|
10
|
Mackels L, Servais L. The Importance of Early Treatment of Inherited Neuromuscular Conditions. J Neuromuscul Dis 2024; 11:253-274. [PMID: 38306060 DOI: 10.3233/jnd-230189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
There has been tremendous progress in treatment of neuromuscular diseases over the last 20 years, which has transformed the natural history of these severely debilitating conditions. Although the factors that determine the response to therapy are many and in some instance remain to be fully elucidated, early treatment clearly has a major impact on patient outcomes across a number of inherited neuromuscular conditions. To improve patient care and outcomes, clinicians should be aware of neuromuscular conditions that require prompt treatment initiation. This review describes data that underscore the importance of early treatment of children with inherited neuromuscular conditions with an emphasis on data resulting from newborn screening efforts.
Collapse
Affiliation(s)
- Laurane Mackels
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Adult Neurology Department, Citadelle Hospital, Liège, Belgium
| | - Laurent Servais
- Neuromuscular Centre, Division of Paediatrics, University and University Hospital of Liège, Liège, Belgium
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford & NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
11
|
Kastreva K, Chamova T, Blagoeva S, Bichev S, Mihaylova V, Meyer S, Thompson R, Cherninkova S, Guergueltcheva V, Lochmuller H, Tournev I. Characterization of Clinical Phenotypes in Congenital Myasthenic Syndrome Associated with the c.1327delG Frameshift Mutation in CHRNE Encoding the Acetylcholine Receptor Epsilon Subunit. J Neuromuscul Dis 2024; 11:1011-1020. [PMID: 38995797 PMCID: PMC11380250 DOI: 10.3233/jnd-230235] [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] [Accepted: 06/10/2024] [Indexed: 07/14/2024]
Abstract
Background Congenital myasthenic syndromes (CMS) are a group of rare but often treatable inherited disorders of neuromuscular transmission characterized by fatigable skeletal muscle weakness. In this paper we present the largest phenotypic analysis to date of a cohort of patients carrying the pathogenic variant c.1327delG in the CHRNE gene, leading to CHRNE-CMS. Objective This study aims to identify the phenotypic variability in CMS associated with c.1327delG mutation in the CHRNE gene. Methods Disease specific symptoms were assessed using specific standardized tests for autoimmune myasthenia (Quantitative Myasthenia Gravis score) as well as patient-reported scales for symptom severity. Evaluated clinical manifestations included ocular symptoms (ophthalmoparesis and ptosis), bulbar weakness, axial muscle weakness, proximal and distal muscle weakness, and respiratory function. Patients were allocated into three groups according to clinical impression of disease severity: mild, moderate, and severe. Results We studied 91 Bulgarian Roma patients, carrying the same causative homozygous CHRNE c.1327delG mutation. Bulbar weakness was present in patients throughout all levels of severity of CHRNE-CMS in this study. However, difficulties in eating and swallowing are more prominent characteristics in the moderate and severe clinical phenotypes. Diplopia and ptosis resulting from fatigue of the extraocular muscles were permanent features regardless of disease severity or age. Levels of axial, proximal and distal muscle weakness were variable between disease groups. The statistical analysis showed significant differences between the patients in the three groups, emphasizing a possible variation in symptom manifestation in the evaluated patient population despite the disease originating from the same genetic mutation. Impairment of respiratory function was more prominent in severely affected patients, which might result from loss of compensatory muscle function in those individuals. Conclusion Results from our study indicate significant phenotypic heterogeneity leading to mild, moderate, or severe clinical manifestation in CHRNE-CMS, despite the genotypic homogeneity.
Collapse
Affiliation(s)
- Kristina Kastreva
- Department of Neurology, Expert Centre for Hereditary Neurologic and Metabolic Disorders, University Hospital “Alexandrovska”, Sofia, Bulgaria
- Medical University Sofia, Sofia, Bulgaria
| | - Teodora Chamova
- Department of Neurology, Expert Centre for Hereditary Neurologic and Metabolic Disorders, University Hospital “Alexandrovska”, Sofia, Bulgaria
- Medical University Sofia, Sofia, Bulgaria
| | - Stanislava Blagoeva
- Department of Neurology, Expert Centre for Hereditary Neurologic and Metabolic Disorders, University Hospital “Alexandrovska”, Sofia, Bulgaria
| | - Stoyan Bichev
- National Genetics Laboratory, University Hospital of Obstetrics and Gynecology “Maichin Dom”
| | | | - Stefanie Meyer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Rachel Thompson
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Sylvia Cherninkova
- Department of Neurology, Expert Centre for Hereditary Neurologic and Metabolic Disorders, University Hospital “Alexandrovska”, Sofia, Bulgaria
| | - Velina Guergueltcheva
- Department of Neurology, University Hospital “SofiaMed”, Sofia, Bulgaria
- Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Hanns Lochmuller
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - Ivailo Tournev
- Department of Neurology, Expert Centre for Hereditary Neurologic and Metabolic Disorders, University Hospital “Alexandrovska”, Sofia, Bulgaria
- Medical University Sofia, Sofia, Bulgaria
- Department of Cognitive Science and Psychology, New Bulgarian University, Sofia, Bulgaria
| |
Collapse
|
12
|
Chen X, Qiu J, Gao Z, Liu B, Zhang C, Yu W, Yang J, Shen Y, Qi L, Yao X, Sun H, Yang X. Myasthenia gravis: Molecular mechanisms and promising therapeutic strategies. Biochem Pharmacol 2023; 218:115872. [PMID: 37865142 DOI: 10.1016/j.bcp.2023.115872] [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: 09/03/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Myasthenia gravis (MG) is a type of autoimmune disease caused by the blockage of neuromuscular junction transmission owing to the attack of autoantibodies on transmission-related proteins. Related antibodies, such as anti-AChR, anti-MuSK and anti-LRP4 antibodies, can be detected in most patients with MG. Although traditional therapies can control most symptoms, several challenges remain to be addressed, necessitating the development of more effective and safe treatment strategies for MG. With the in-depth exploration on the mechanism and immune targets of MG, effective therapies, especially therapies using biologicals, have been reported recently. Given the important roles of immune cells, cytokines and intercellular interactions in the pathological process of MG, B-cell targeted therapy, T-cell targeted therapy, proteasome inhibitors targeting plasma cell, complement inhibitors, FcRn inhibitors have been developed for the treatment of MG. Although these novel therapies exert good therapeutic effects, they may weaken the immunity and increase the risk of infection in MG patients. This review elaborates on the pathogenesis of MG and discusses the advantages and disadvantages of the strategies of traditional treatment and biologicals. In addition, this review emphasises that combined therapy may have better therapeutic effects and reducing the risk of side effects of treatments, which has great prospects for the treatment of MG. With the deepening of research on immunotherapy targets in MG, novel opportunities and challenges in the treatment of MG will be introduced.
Collapse
Affiliation(s)
- Xin Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Jiayi Qiu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Zihui Gao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Chen Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Weiran Yu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Jiawen Yang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Xinlei Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| |
Collapse
|
13
|
Qaisar R. Targeting neuromuscular junction to treat neuromuscular disorders. Life Sci 2023; 333:122186. [PMID: 37858716 DOI: 10.1016/j.lfs.2023.122186] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
The integrity and preservation of the neuromuscular junction (NMJ), the interface between the motor neuron and skeletal muscle, is critical for maintaining a healthy skeletal muscle. The structural and/or functional defects in the three cellular components of NMJ, namely the pre-synaptic terminal, synaptic cleft, and post-synaptic region, negatively affect skeletal muscle mass and/or strength. Therefore, NMJ repair appears to be an appropriate therapy for muscle disorders. Mouse models provide a detailed molecular characterization of various cellular components of NMJ with relevance to human diseases. This review discusses different molecular targets on the three cellular components of NMJ for treating muscle diseases. The potential effects of these therapies on NMJ morphology and motor performance, their therapeutic efficacy, and clinical relevance are discussed. Collectively, the available data supports targeting NMJ alone or as an adjunct therapy in treating muscle disorders. However, the potential impact of such interventions on human patients with muscle disorders requires further investigation.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Space Medicine Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Cardiovascular Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
14
|
Almatrafi AM, Alluqmani MM, Basit S. Homozygous Duplication in the CHRNE in a Family with Congenital Myasthenic Syndrome 4C: 18-Year Follow Up. Biomedicines 2023; 11:2983. [PMID: 38001983 PMCID: PMC10668953 DOI: 10.3390/biomedicines11112983] [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: 09/11/2023] [Revised: 10/29/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Congenital myasthenic syndromes (CMSs) are rare inherited diseases characterized by muscle weakness and fatigability on exertion resulting from defects in the neuromuscular junctions. Mutations in 32 genes have been reported as the underlying causes of CMS, with mutations in the cholinergic receptor nicotinic epsilon subunit (CHRNE) being the most common cause of the disease. Methodology and Materials: This study investigated a large consanguineous family with multiple individuals suffering from abnormal fatigue and muscle weakness in the ocular and limb regions. Moreover, the affected individuals were followed up for 18 years to observe the clinical course of the disease. RESULTS High-quality exome sequencing followed by bidirectional Sanger sequencing revealed a homozygous duplication variant (NM_000080.4: c.1220-8_1227dup) in the splice acceptor site of exon 11 of the CHRNE gene. This variant is predicted to cause frameshift and premature termination (p.Cys410ProfsTer51). Both parents had heterozygous duplication variants with no clinical symptoms. The personalized treatment of the affected individuals resulted in a marked improvement in the clinical symptoms. More than 80% of the disease symptoms in the affected individuals subsided after the use of pyridostigmine and salbutamol (4 mg). CONCLUSIONS This is the first report of long-term follow up of cases with homozygous insertion (c.1220-8_1227dup) in the CHRNE gene. Furthermore, this report expands the phenotypic symptoms associated with the CHRNE mutation.
Collapse
Affiliation(s)
- Ahmad M. Almatrafi
- Department of Biology, College of Science, Taibah University, Medina 42353, Saudi Arabia;
| | - Majed M. Alluqmani
- Department of Neurology, College of Medicine, Taibah University, Medina 42353, Saudi Arabia;
| | - Sulman Basit
- Department of Biochemistry and Molecular Medicine, College of Medicine, Taibah University, Medina 42353, Saudi Arabia
| |
Collapse
|
15
|
Orriëns LB, Eker D, Braakman HMH, Merkus PJFM, Erasmus CE. Recognising symptoms of congenital myasthenic syndromes in children: A guide for paediatricians. Acta Paediatr 2023; 112:2434-2439. [PMID: 37551152 DOI: 10.1111/apa.16945] [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: 04/11/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/09/2023]
Abstract
AIM Congenital myasthenic syndromes (CMS) are a rare and diverse group of treatable neuromuscular transmission disorders. Diagnosis is often substantially delayed. This study aimed to identify common symptoms of CMS in children and their manifestation to aid diagnosis and early intervention. METHODS We performed a retrospective cohort study, including 18 children (median age 13 years, range 9 years 5 months-18 years 0 month) with CMS. Data on CMS symptoms and their manifestation were extracted from patients' charts and supplemented with parental telephone interviews. Descriptive analyses were used to identify common symptoms. RESULTS A median diagnostic delay of 4 years and 7 months (interquartile range: 51 months) was observed. Proximal muscle weakness (100%), ptosis (89%), clumsy gait (82%), difficulty eating solid foods (78%) and recurrent respiratory tract infections (72%) were most common in these patients. Symptoms mostly co-occurred and frequently had a fluctuating character, aggravated by infections or fatigue. CONCLUSION Early referral to diagnose CMS is crucial to enable timely initiation of treatment. Heightened attention to a combination of symptoms related to muscle weakness, rather than individual symptoms, should support paediatricians in flagging these neuromuscular disorders. Medical history taking should be tailored to parents' perceptions, asking questions about recognisable symptoms of muscle weakness.
Collapse
Affiliation(s)
- Lynn B Orriëns
- Division of Paediatrics, Department of Paediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Dilan Eker
- Division of Paediatrics, Department of Paediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Hilde M H Braakman
- Division of Paediatrics, Department of Paediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Peter J F M Merkus
- Division of Paediatrics, Department of Paediatric Pulmonology, Radboud University Medical Centre, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Corrie E Erasmus
- Division of Paediatrics, Department of Paediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Amalia Children's Hospital, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Mishra S, Nair KV, Shukla A. Delineation of molecular characteristics of congenital myasthenic syndromes in Indian families and review of literature. Clin Dysmorphol 2023; 32:162-167. [PMID: 37646703 DOI: 10.1097/mcd.0000000000000465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Congenital myasthenic syndromes (CMS) are rare, heterogeneous, and often treatable genetic disorders depending on the underlying molecular defect. We performed a detailed clinical evaluation of seven patients from five unrelated families. Exome sequencing was performed on five index patients. Clinically significant variants were identified in four CMS disease-causing genes: COLQ (3/7), CHRNE (2/7), DOK7 (1/7), and RAPSN (1/7). We identified two novel variants, c.930_933delCATG in DOK7 and c.1016_1032 + 2dup in CHRNE . A common pathogenic variant, c.955-2A>C, has been identified in COLQ -related CMS patients. Homozygosity mapping of this COLQ variant in patients from two unrelated families revealed that it was located in a common homozygous region of 3.2 Mb on chromosome 3 and was likely to be inherited from a common ancestor. Patients with COLQ variants had generalized muscle weakness, those with DOK7 and RAPSN variants had limb-girdle weakness, and those with CHRNE variants had predominant ocular weakness. Patients with COLQ and DOK7 variants showed improvement with salbutamol and CHRNE with pyridostigmine therapy. This study expands the mutational spectrum and adds a small but significant cohort of CMS patients from India. We also reviewed the literature to identify genetic subtypes of CMS in India.
Collapse
Affiliation(s)
- Shivani Mishra
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | | |
Collapse
|
17
|
Spendiff S, Dong Y, Maggi L, Rodríguez Cruz PM, Beeson D, Lochmüller H. 260th ENMC International Workshop: Congenital myasthenic syndromes 11-13 March 2022, Hoofddorp, The Netherlands. Neuromuscul Disord 2023; 33:111-118. [PMID: 36609117 DOI: 10.1016/j.nmd.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Sally Spendiff
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Yin Dong
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Pedro M Rodríguez Cruz
- Centro Nacional de Análisis Genómico (CNAG-CRG), Centre for Genomic Regulation, Barcelona, Spain; Department of Human Genetics, Université Cheikh Anta Diop, Dakar, Senegal; Department of Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada; Department of Medicine, Division of Neurology, The Ottawa Hospital, Ottawa, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada; Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.
| |
Collapse
|
18
|
Masingue M, Cattaneo O, Wolff N, Buon C, Sternberg D, Euchparmakian M, Boex M, Behin A, Mamchaouhi K, Maisonobe T, Nougues MC, Isapof A, Fontaine B, Messéant J, Eymard B, Strochlic L, Bauché S. New mutation in the β1 propeller domain of LRP4 responsible for congenital myasthenic syndrome associated with Cenani-Lenz syndrome. Sci Rep 2023; 13:14054. [PMID: 37640745 PMCID: PMC10462681 DOI: 10.1038/s41598-023-41008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023] Open
Abstract
Congenital myasthenic syndromes (CMS) are a clinically and genetically heterogeneous group of rare diseases due to mutations in neuromuscular junction (NMJ) protein-coding genes. Until now, many mutations encoding postsynaptic proteins as Agrin, MuSK and LRP4 have been identified as responsible for increasingly complex CMS phenotypes. The majority of mutations identified in LRP4 gene causes bone diseases including CLS and sclerosteosis-2 and rare cases of CMS with mutations in LRP4 gene has been described so far. In the French cohort of CMS patients, we identified a novel LRP4 homozygous missense mutation (c.1820A > G; p.Thy607Cys) within the β1 propeller domain in a patient presenting CMS symptoms, including muscle weakness, fluctuating fatigability and a decrement in compound muscle action potential in spinal accessory nerves, associated with congenital agenesis of the hands and feet and renal malformation. Mechanistic expression studies show a significant decrease of AChR aggregation in cultured patient myotubes, as well as altered in vitro binding of agrin and Wnt11 ligands to the mutated β1 propeller domain of LRP4 explaining the dual phenotype characterized clinically and electoneuromyographically in the patient. These results expand the LRP4 mutations spectrum associated with a previously undescribed clinical association involving impaired neuromuscular transmission and limb deformities and highlighting the critical role of a yet poorly described domain of LRP4 at the NMJ. This study raises the question of the frequency of this rare neuromuscular form and the future diagnosis and management of these cases.
Collapse
Affiliation(s)
- Marion Masingue
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
- Service de Neuromyologie, Centre de Référence Neuromusculaire, APHP, Paris, France
| | - Olivia Cattaneo
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Nicolas Wolff
- Institut Pasteur, Channel Receptors Unit, UMR CNRS 3571, Université de Paris, Paris, France
| | - Céline Buon
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
| | - Damien Sternberg
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
- Service de Biochimie Métabolique, UF Cardiogenetics and Myogenetics, Hôpital de la Pitié-Salpêtrière, APHP, Paris, France
| | - Morgane Euchparmakian
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
| | - Myriam Boex
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
| | - Anthony Behin
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
- Service de Neuromyologie, Centre de Référence Neuromusculaire, APHP, Paris, France
| | - Kamel Mamchaouhi
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
| | - Thierry Maisonobe
- Département de Neurophysiologie Clinique, Centre de Référence des Pathologies Neuromusculaires, Hôpital de la Pitié-Salpêtrière, APHP, Paris, France
| | - Marie-Christine Nougues
- Département de Neuropédiatrie, Centre de Référence des Pathologies Neuromusculaires, Hôpital Trousseau, APHP, Paris, France
| | - Arnaud Isapof
- Département de Neuropédiatrie, Centre de Référence des Pathologies Neuromusculaires, Hôpital Trousseau, APHP, Paris, France
| | - Bertrand Fontaine
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
- Service de Neuromyologie, Centre de Référence Canalopathie, Hôpital de la Pitié-Salpêtrière, APHP, Paris, France
| | - Julien Messéant
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
| | - Bruno Eymard
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
| | - Laure Strochlic
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France
| | - Stéphanie Bauché
- INSERM, Myology Research Center-UMRS974, Hôpital Universitaire de la Pitié-Salpêtrière, Institut de Myologie, Sorbonne Université, 105 Boulevard de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
19
|
Horita H, Sato I, Ju H, Lee E, Ikeda M, Kuwahara S, Habata Y. 14- and 17-Membered Macrocycles Containing Amide, Amino, and Carbamate Groups in The Monocyclic Skeleton: An Accidental Byproduct Obtained from a Residue after Separation. J Org Chem 2023. [PMID: 37184952 DOI: 10.1021/acs.joc.2c02733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
A 14-membered cyclic compound (3) containing amide, amino, and carbamate groups, which was serendipitously obtained in the oily residue after the separation of 4-benzyl-1,4,7,10-tetraazacyldododecane-2,6-dione (2a) and 4,16-dibenzyl-1,4,7,10,13,16,19,22-octaazacyclo-tetracosane-2,6,14,18-tetraone (2b), is reported. The structure of 3 is formally a CO2 insertion between positions 3 and 4 of the 12-membered ring in 2a. The CO2 insertion was confirmed in the synthesis of diethyl 2,2'-(benzylazanediyl)diacetate (1) by the reaction of benzylamine with ethyl bromoacetate using K2CO3 as the base. In addition, the selective synthesis of 3 and ethyl N-benzyl-N-((2-ethoxy-2-oxoethoxy)carbonyl)glycinate (5) and their kinetic behavior are reported. The reaction of 5 with triethylenetetramine afforded a 17-membered macrocycle (7), which was obtained in an 18% yield. Compounds 6 and 8 were prepared from 3 and 7 by introducing benzyl groups to improve their solubility in organic solvents. Titration experiments using 1H NMR showed that both 6 and 8 exhibit Li+ selectivity.
Collapse
Affiliation(s)
| | | | - Huiyeong Ju
- Western Seoul Center, Korea Basic Science Institute, 150 Bugahyeon-ro, Seodaemun-gu, Seoul 03759, Republic of Korea
| | - Eunji Lee
- Department of Chemistry, Gangneung-Wonju National University, Gangneung 25457, South Korea
| | - Mari Ikeda
- Education Center, Faculty of Engineering, Chiba Institute of Technology, 2-1-1 Shibazono, Narashino, Chiba 275-0023, Japan
| | | | | |
Collapse
|
20
|
Horibe T, Shimomura H, Tokunaga S, Taniguchi N, Lee T, Kimura S, Takeshima Y. A Pediatric Case of COLQ-Related Congenital Myasthenic Syndrome with Marked Fatigue. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10050769. [PMID: 37238317 DOI: 10.3390/children10050769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
Congenital myasthenic syndrome (CMS) is a clinically and genetically heterogeneous inherited disorder that is treatable. Although the disease usually develops at birth or during infancy, some patients develop the disease in the second to third decades of life. Collagen-like tail subunit of asymmetric acetylcholinesterase (COLQ)-related CMS is CMS with mutations in the COLQ, which results in end-plate acetylcholinesterase deficiency. Diagnostic delay is common in patients with later-onset CMS due to slow progression and fluctuating symptoms. Understanding CMS with atypical and unusual presentations is important to treat this condition effectively. Here, we report a case of COLQ-related CMS. A 10-year-old girl presented with only marked fatigue, which was provoked by exercise but improved after 30-60 min of rest. While motor nerve conduction velocity was normal, a compound muscle action potential (CMAP) with four peaks was recorded. Repetitive stimulation of the accessory nerve exhibited a decrease in CMAP amplitude. Genetic tests revealed compound heterozygous mutations in COLQ (c.1196-1_1197delinsTG and c.1354C>T). Treatment with salbutamol improved fatigue but not the electrophysiological markers. Thus, significant fatigue is a hallmark of COLQ-related CMS; early diagnosis is essential for ensuring appropriate treatment.
Collapse
Affiliation(s)
- Takuya Horibe
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya 663-8501, Japan
| | - Hideki Shimomura
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya 663-8501, Japan
| | - Sachi Tokunaga
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya 663-8501, Japan
| | - Naoko Taniguchi
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya 663-8501, Japan
| | - Tomoko Lee
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya 663-8501, Japan
| | - Shigemi Kimura
- Children's Rehabilitation, Sleep and Development Medical Center, Hyogo Prefectural Rehabilitation Central Hospital, Kobe 651-2134, Japan
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo Medical University School of Medicine, Nishinomiya 663-8501, Japan
| |
Collapse
|
21
|
Vlckova M, Prchalova D, Zimmermann P, Haberlova J, Bendova S, Moslerova V, Stranecky V, Sedlacek Z, Hancarova M. A new patient with congenital myasthenic syndrome type 20 due to compound heterozygous missense SLC5A7 variants suggests trends in genotype-phenotype correlation. Mol Genet Genomic Med 2023:e2154. [PMID: 36840359 DOI: 10.1002/mgg3.2154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Congenital myasthenic syndromes (CMSs) are characterized by hypotonia, episodic apnea, muscle weakness, ptosis and generalized fatigability. CMS type 20 (CMS20) is a rare disorder caused by variants in SLC5A7. In contrast to most other CMSs, CMS20 is also associated with neurodevelopmental disorders (NDDs). Only 19 patients from 14 families have been reported so far. METHODS We studied a 12-year-old boy with symptoms manifested at six weeks of age. Later, he also showed speech delay, moderate intellectual disability and autism. Analysis of CMS genes known at the time of clinical diagnosis yielded no results. Trio exome sequencing (ES) was performed. RESULTS ES revealed compound heterozygosity for two SLC5A7 variants, p.(Asn431Lys) and p.(Ile291Thr). While the first variant was absent from all databases, the second variant has already been described in one patient. In silico analysis of known pathogenic SLC5A7 variants showed that variants with a higher predicted deleteriousness may be associated with earlier onset and increased severity of neuromuscular manifestations. CONCLUSION Our patient confirms that CMS20 can be associated with NDDs. The study illustrates the strength of ES in deciphering the genetic basis of rare diseases, contributes to characterization of CMS20 and suggests trends in genotype-phenotype correlation in CMS20.
Collapse
Affiliation(s)
- Marketa Vlckova
- Department of Biology and Medical Genetics, Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Darina Prchalova
- Department of Biology and Medical Genetics, Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Pavel Zimmermann
- Department of Statistics and Probability, Faculty of Informatics and Statistics, University of Economics, Prague, Czech Republic
| | - Jana Haberlova
- Department of Pediatric Neurology, Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Sarka Bendova
- Department of Biology and Medical Genetics, Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Veronika Moslerova
- Department of Biology and Medical Genetics, Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Viktor Stranecky
- Department of Pediatrics and Adolescent Medicine, Diagnostic and Research Unit for Rare Diseases, Charles University First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Zdenek Sedlacek
- Department of Biology and Medical Genetics, Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Miroslava Hancarova
- Department of Biology and Medical Genetics, Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
22
|
Rodríguez Cruz PM, Ravenscroft G, Natera D, Carr A, Manzur A, Liu WW, Vella NR, Jericó I, Gonzalez-Quereda L, Gallano P, Montalto SA, Davis MR, Lamont PJ, Laing NG, Bourque P, Nascimento A, Muntoni F, Polavarapu K, Lochmüller H, Palace J, Beeson D. A novel phenotype of AChR-deficiency syndrome with predominant facial and distal weakness resulting from the inclusion of an evolutionary alternatively-spliced exon in CHRNA1. Neuromuscul Disord 2023; 33:161-168. [PMID: 36634413 DOI: 10.1016/j.nmd.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Primary acetylcholine receptor deficiency is the most common subtype of congenital myasthenic syndrome, resulting in reduced amount of acetylcholine receptors expressed at the muscle endplate and impaired neuromuscular transmission. AChR deficiency is caused mainly by pathogenic variants in the ε-subunit of the acetylcholine receptor encoded by CHRNE, although pathogenic variants in other subunits are also seen. We report the clinical and molecular features of 13 patients from nine unrelated kinships with acetylcholine receptor deficiency harbouring the CHRNA1 variant NM_001039523.3:c.257G>A (p.Arg86His) in homozygosity or compound heterozygosity. This variant results in the inclusion of an alternatively-spliced evolutionary exon (P3A) that causes expression of a non-functional acetylcholine receptor α-subunit. We compare the clinical findings of this group to the other cases of acetylcholine receptor deficiency within our cohort. We report differences in phenotype, highlighting a predominant pattern of facial and distal weakness in adulthood, predominantly in the upper limbs, which is unusual for acetylcholine receptor deficiency syndromes, and more in keeping with slow-channel syndrome or distal myopathy. Finally, we stress the importance of including alternative exons in variant analysis to increase the probability of achieving a molecular diagnosis.
Collapse
Affiliation(s)
- Pedro M Rodríguez Cruz
- CNAG-CRG, Centro Nacional de Análisis Genómico - Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Gianina Ravenscroft
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia; Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - Daniel Natera
- Neuromuscular Unit, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Aisling Carr
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, NIHR Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Wei Wei Liu
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, UK
| | - Norbert R Vella
- Department of Neuroscience, Mater Dei Hospital, Msida, Malta
| | - Ivonne Jericó
- Department of Neurology, Hospital Universitario de Navarra, IdisNa (Instituto Investigación Sanitaria Navarra), Pamplona, Spain
| | - Lidia Gonzalez-Quereda
- Center for the Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain; Department of Genetics, Hospital de Sant Pau, IIB Sant Pau, Barcelona, Spain
| | - Pia Gallano
- Center for the Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain; Department of Genetics, Hospital de Sant Pau, IIB Sant Pau, Barcelona, Spain
| | | | - Mark R Davis
- Neurogenetic Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, Western Australian Department of Health, Nedlands, WA, Australia
| | - Phillipa J Lamont
- Department of Neurology, Royal Perth Hospital, Nedlands, WA, Australia
| | - Nigel G Laing
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia; Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia; Neurogenetic Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, Western Australian Department of Health, Nedlands, WA, Australia
| | - Pierre Bourque
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada
| | | | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kiran Polavarapu
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada
| | - Hanns Lochmüller
- CNAG-CRG, Centro Nacional de Análisis Genómico - Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain; Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada; Division of Neurology, Department of Medicine, The Ottawa Hospital; and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada; Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany.
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - David Beeson
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Krenn M, Sener M, Rath J, Zulehner G, Keritam O, Wagner M, Laccone F, Iglseder S, Marte S, Baumgartner M, Eisenkölbl A, Liechtenstein C, Rudnik S, Quasthoff S, Grinzinger S, Spenger J, Wortmann SB, Löscher WN, Zimprich F, Kellersmann A, Rappold M, Bernert G, Freilinger M, Cetin H. The clinical and molecular landscape of congenital myasthenic syndromes in Austria: a nationwide study. J Neurol 2023; 270:909-916. [PMID: 36308527 PMCID: PMC9886627 DOI: 10.1007/s00415-022-11440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Congenital myasthenic syndromes (CMS) are a heterogeneous group of disorders caused by genetic defects resulting in impaired neuromuscular transmission. Although effective treatments are available, CMS is probably underdiagnosed, and systematic clinico-genetic investigations are warranted. METHODS We used a nationwide approach to collect Austrian patients with genetically confirmed CMS. We provide a clinical and molecular characterization of this cohort and aimed to ascertain the current frequency of CMS in Austria. RESULTS Twenty-eight cases with genetically confirmed CMS were identified, corresponding to an overall prevalence of 3.1 per million (95% CI 2.0-4.3) in Austria. The most frequent genetic etiology was CHRNE (n = 13), accounting for 46.4% of the cohort. Within this subgroup, the variant c.1327del, p.(Glu443Lysfs*64) was detected in nine individuals. Moreover, causative variants were found in DOK7 (n = 4), RAPSN (n = 3), COLQ (n = 2), GMPPB (n = 2), CHAT (n = 1), COL13A1 (n = 1), MUSK (n = 1) and AGRN (n = 1). Clinical onset within the first year of life was reported in one half of the patients. Across all subtypes, the most common symptoms were ptosis (85.7%), lower limb (67.9%), upper limb (60.7%) and facial weakness (60.7%). The majority of patients (96.4%) received specific treatment, including acetylcholinesterase inhibitors in 20, adrenergic agonists in 11 and 3,4-diaminopyridine in nine patients. CONCLUSIONS Our study presents the first systematic characterization of individuals with CMS in Austria, providing prevalence estimates and genotype-phenotype correlations that may help to improve the diagnostic approach and patient management.
Collapse
Affiliation(s)
- Martin Krenn
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Merve Sener
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Jakob Rath
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Gudrun Zulehner
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Omar Keritam
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Matias Wagner
- Institute of Human Genetics, Technical University of Munich, Munich, Germany.,Institute for Neurogenomics, Helmholtz Center Munich, Munich, Germany
| | - Franco Laccone
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Stephan Iglseder
- Department of Neurology, Krankenhaus Barmherzige Brüder, Linz, Austria
| | | | - Manuela Baumgartner
- Department of Neuropaediatrics, Hospital Barmherzige Schwestern Linz, Linz, Austria
| | - Astrid Eisenkölbl
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Christian Liechtenstein
- Department of Paediatrics and Adolescent Medicine, Villach Regional Hospital, Villach, Austria
| | - Sabine Rudnik
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Quasthoff
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Susanne Grinzinger
- Department of Neurology, Salzburger Landeskliniken, Paracelsus Medical University, Salzburg, Austria
| | - Johannes Spenger
- University Children's Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Saskia B Wortmann
- University Children's Hospital, Paracelsus Medical University, Salzburg, Austria.,Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Wolfgang N Löscher
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Fritz Zimprich
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Mika Rappold
- Department of Pediatrics, Klinik Favoriten, Vienna, Austria
| | | | - Michael Freilinger
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Hakan Cetin
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Chan C, Emery L, Maltese C, Kumar A, Aliu E, Naik S, Paul D. A Novel Homozygous Variant in the CHRNE Gene in 2 Siblings with Congenital Myasthenic Syndrome. Child Neurol Open 2023; 10:2329048X231216432. [PMID: 38034490 PMCID: PMC10685742 DOI: 10.1177/2329048x231216432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/20/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
Cholinergic receptor nicotinic epsilon (CHRNE) subunit mutations cause postsynaptic type of congenital myasthenic syndrome either as a primary acetylcholine-receptor deficiency or abnormal channel kinetics in the receptor. We report a novel homozygous variant (c.322C > T, p.Pro108Ser) in the epsilon subunit causing primary acetylcholine-receptor deficiency in two siblings. Two siblings presented with fatigable weakness. Both siblings had whole exome sequencing showing a homozygous variant (c.322C > T, p.Pro108Ser) of unknown significance in the epsilon subunit. Electromyography/nerve conduction study with repetitive nerve stimulation on one sibling showed a defect in neuromuscular junction transmission. Pseudoephedrine and fluoxetine for suspected slow-channel congenital myasthenic syndrome yielded no improvement. A trial of pyridostigmine led to clinical improvement. Given the clinical presentation, consanguinity, homozygous genetic variant, and response to pyridostigmine, we rationalize the homozygous variant (c.322C > T, p.Pro108Ser) in cholinergic receptor nicotinic epsilon subunit causes the primary acetylcholine-receptor deficiency congenital myasthenic syndrome.
Collapse
Affiliation(s)
- Cassie Chan
- Department of Neurology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Lucy Emery
- Penn State Health College of Medicine, Hershey, PA, USA
| | | | - Ashutosh Kumar
- Department of Neurology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pediatrics, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Ermal Aliu
- Department of Genetics, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Sunil Naik
- Department of Neurology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pediatrics, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Dustin Paul
- Department of Neurology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pediatrics, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
25
|
Stojkovic T, Masingue M, Turmel H, Hezode-Arzel M, Béhin A, Leonard-Louis S, Bassez G, Bauché S, Blondy P, Richard P, Sternberg D, Eymard B, Fournier E, Villar-Quiles RN. Diagnostic yield of a practical electrodiagnostic protocol discriminating between different congenital myasthenic syndromes. Neuromuscul Disord 2022; 32:870-878. [PMID: 36522822 DOI: 10.1016/j.nmd.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022]
Abstract
Congenital myasthenic syndromes (CMS) are a group of heterogeneous diseases of the neuromuscular junction. We report electrodiagnostic testing (EDX) and genetic findings in a series of 120 CMS patients tested with a simple non-invasive EDX workup with surface recording of CMAPs and 3Hz repetitive nerve stimulation of accessory, radial and deep fibular nerves. Five ENMG phenotypes were retrieved based on the presence or not of R-CMAPs and the distribution pattern of decremental CMAP responses which significantly correlated with genetic findings (p <0.00001). R-CMAPs were found in all COLQ-mutated patients (CMS1A) and Slow Channel CMS (SCCMS) (CMS1B). CMS1A exhibited greater decrements in accessory nerve RNS than CMS1B. Patients without R-CMAPs were classified into CMS2A (DOK7-, MUSK-, GFPT1-, GMPPB-, TOR1AIP-mutated) when exhibiting predominant accessory nerve RNS decrements, CMS2B (CHRNE, CHRND, RAPSN) with predominant radial nerve RNS decrements, or CMS2C (AGRN) if there were predominant fibular decrements. Our algorithm may have a major impact on diagnostic and therapeutic monitoring in CMS patients, as well as for validation of the pathogenicity of genetic variants. It should also be part of the evaluation of unexplained muscle weakness or complex neuromuscular phenotypes.
Collapse
Affiliation(s)
- Tanya Stojkovic
- Reference Center for Neuromuscular Disorders (Nord/Est/Ile de France), Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Pitié-Salpêtrière Hospital, Paris, France; Centre de Recherche en Myologie, Sorbonne Université-Inserm UMRS974, Paris, France
| | - Marion Masingue
- Reference Center for Neuromuscular Disorders (Nord/Est/Ile de France), Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Pitié-Salpêtrière Hospital, Paris, France
| | - Helène Turmel
- Department of Neurophysiology, APHP, Pitié Salpetrière hospital, Paris, France
| | | | - Anthony Béhin
- Reference Center for Neuromuscular Disorders (Nord/Est/Ile de France), Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Pitié-Salpêtrière Hospital, Paris, France
| | - Sarah Leonard-Louis
- Reference Center for Neuromuscular Disorders (Nord/Est/Ile de France), Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Pitié-Salpêtrière Hospital, Paris, France
| | - Guillaume Bassez
- Reference Center for Neuromuscular Disorders (Nord/Est/Ile de France), Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Pitié-Salpêtrière Hospital, Paris, France; Centre de Recherche en Myologie, Sorbonne Université-Inserm UMRS974, Paris, France
| | - Stéphanie Bauché
- Centre de Recherche en Myologie, Sorbonne Université-Inserm UMRS974, Paris, France
| | - Patricia Blondy
- National Reference Center for Muscle Channelopathies, APHP, Pitié Salpetrière hospital, Paris, France; Biochemistry Department, Center of Molecular and Cellular Genetics, APHP, Pitié Salpetrière hospital, Paris, France
| | - Pascale Richard
- Biochemistry Department, Center of Molecular and Cellular Genetics, APHP, Pitié Salpetrière hospital, Paris, France
| | - Damien Sternberg
- National Reference Center for Muscle Channelopathies, APHP, Pitié Salpetrière hospital, Paris, France; Biochemistry Department, Center of Molecular and Cellular Genetics, APHP, Pitié Salpetrière hospital, Paris, France
| | - Bruno Eymard
- Reference Center for Neuromuscular Disorders (Nord/Est/Ile de France), Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Pitié-Salpêtrière Hospital, Paris, France
| | - Emmanuel Fournier
- Department of Neurophysiology, APHP, Pitié Salpetrière hospital, Paris, France; National Reference Center for Muscle Channelopathies, APHP, Pitié Salpetrière hospital, Paris, France; Department of Physiology, Sorbonne University, Faculté de médecine Pitié-Salpêtrière, Paris, France
| | - Rocío Nur Villar-Quiles
- Reference Center for Neuromuscular Disorders (Nord/Est/Ile de France), Institute of Myology, Pitié-Salpêtrière Hospital, APHP, Pitié-Salpêtrière Hospital, Paris, France; Centre de Recherche en Myologie, Sorbonne Université-Inserm UMRS974, Paris, France
| |
Collapse
|
26
|
El Kadiri Y, Ratbi I, Sefiani A, Lyahyai J. Novel copy number variation of COLQ gene in a Moroccan patient with congenital myasthenic syndrome: a case report and review of the literature. BMC Neurol 2022; 22:292. [PMID: 35932018 PMCID: PMC9354381 DOI: 10.1186/s12883-022-02822-y] [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: 03/14/2022] [Accepted: 07/29/2022] [Indexed: 11/11/2022] Open
Abstract
Background Congenital myasthenic syndromes (CMSs) are rare genetic diseases due to abnormalities of the neuromuscular junction leading to permanent or transient muscle fatigability and weakness. To date, 32 genes were found to be involved in CMSs with autosomal dominant and/or recessive inheritance patterns. CMS with acetylcholinesterase deficiency, in particular, was determined to be due to biallelic mutations of COLQ gene with early-onset clinical signs. Here, we report clinical features and novel molecular findings of COLQ-related CMS in a Moroccan patient with a review of the literature for this rare form. Case presentation In this study, we report the case of a 28-month-old Moroccan female patient with hypotonia, associated to axial muscle weakness, global motor delay, bilateral ptosis, unilateral partial visual field deficiency with normal ocular motility, and fatigable muscle weakness. Clinical exome sequencing revealed a novel homozygous deletion of exon 13 in COLQ gene, NM_005677.4(COLQ):c.(814+1_815-1)_(954+1_955-1) del p.(Gly272Aspfs*11). This finding was subsequently confirmed by quantitative real-time PCR (qPCR) in the proband and her parents. In silico analysis of protein-protein interaction network by STRING tool revealed that 12 proteins are highly associated to COLQ with an elevated confidence score. Treatment with Salbutamol resulted in clear benefits and recovery. Conclusions This clinical observation illustrates the important place of next-generation sequencing in the precise molecular diagnosis of heterogeneous forms of CMS, the appropriate management and targeted treatment, and genetic counseling of families, with a better characterization of the mutational profile of this rare disease in the Moroccan population.
Collapse
Affiliation(s)
- Youssef El Kadiri
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomics Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 100 Rabat, Morocco. .,Department of Medical Genetics, National Institute of Health, BP 769-Agdal, 10 090, Rabat, Morocco.
| | - Ilham Ratbi
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomics Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 100 Rabat, Morocco
| | - Abdelaziz Sefiani
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomics Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 100 Rabat, Morocco.,Department of Medical Genetics, National Institute of Health, BP 769-Agdal, 10 090, Rabat, Morocco
| | - Jaber Lyahyai
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomics Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 100 Rabat, Morocco
| |
Collapse
|
27
|
Jacquier A, Risson V, Simonet T, Roussange F, Lacoste N, Ribault S, Carras J, Theuriet J, Girard E, Grosjean I, Le Goff L, Kröger S, Meltoranta J, Bauché S, Sternberg D, Fournier E, Kostera-Pruszczyk A, O’Connor E, Eymard B, Lochmüller H, Martinat C, Schaeffer L. Severe congenital myasthenic syndromes caused by agrin mutations affecting secretion by motoneurons. Acta Neuropathol 2022; 144:707-731. [PMID: 35948834 PMCID: PMC9468088 DOI: 10.1007/s00401-022-02475-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 01/28/2023]
Abstract
Congenital myasthenic syndromes (CMS) are predominantly characterized by muscle weakness and fatigability and can be caused by a variety of mutations in genes required for neuromuscular junction formation and maintenance. Among them, AGRN encodes agrin, an essential synaptic protein secreted by motoneurons. We have identified severe CMS patients with uncharacterized p.R1671Q, p.R1698P and p.L1664P mutations in the LG2 domain of agrin. Overexpression in primary motoneurons cultures in vitro and in chick spinal motoneurons in vivo revealed that the mutations modified agrin trafficking, leading to its accumulation in the soma and/or in the axon. Expression of mutant agrins in cultured cells demonstrated accumulation of agrin in the endoplasmic reticulum associated with induction of unfolded protein response (UPR) and impaired secretion in the culture medium. Interestingly, evaluation of the specific activity of individual agrins on AChR cluster formation indicated that when secreted, mutant agrins retained a normal capacity to trigger the formation of AChR clusters. To confirm agrin accumulation and secretion defect, iPS cells were derived from a patient and differentiated into motoneurons. Patient iPS-derived motoneurons accumulated mutant agrin in the soma and increased XBP1 mRNA splicing, suggesting UPR activation. Moreover, co-cultures of patient iPS-derived motoneurons with myotubes confirmed the deficit in agrin secretion and revealed a reduction in motoneuron survival. Altogether, we report the first mutations in AGRN gene that specifically affect agrin secretion by motoneurons. Interestingly, the three patients carrying these mutations were initially suspected of spinal muscular atrophy (SMA). Therefore, in the presence of patients with a clinical presentation of SMA but without mutation in the SMN1 gene, it can be worth to look for mutations in AGRN.
Collapse
Affiliation(s)
- Arnaud Jacquier
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Valérie Risson
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Thomas Simonet
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Florine Roussange
- grid.503216.30000 0004 0618 2124INSERM/UEPS UMR 861, Paris Saclay Université, I-STEM, 91100 Corbeil-Essonnes, France
| | - Nicolas Lacoste
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Shams Ribault
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Service de Médecine Physique et de Réadaptation, Hôpital Henry Gabrielle, Hospices Civils de Lyon, 69230 Saint-Genis-Laval, France
| | - Julien Carras
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Julian Theuriet
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Emmanuelle Girard
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Isabelle Grosjean
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Laure Le Goff
- grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Stephan Kröger
- Department of Physiological Genomics, Biomedical Center, Planegg, Martinsried, Germany
| | - Julia Meltoranta
- Department of Physiological Genomics, Biomedical Center, Planegg, Martinsried, Germany
| | - Stéphanie Bauché
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France
| | - Damien Sternberg
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France ,grid.411439.a0000 0001 2150 9058APHP, UF Cardiogénétique et Myogénétique, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Emmanuel Fournier
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France ,grid.411439.a0000 0001 2150 9058AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France ,grid.462844.80000 0001 2308 1657Département d’Éthique de l’Université et des enseignements de Physiologie de la Faculté de Médecine Pitié-Salpêtrière, 75013 Paris, France
| | - Anna Kostera-Pruszczyk
- grid.13339.3b0000000113287408Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Emily O’Connor
- grid.28046.380000 0001 2182 2255Division of Neurology, Department of Medicine, Children’s Hospital of Eastern Ontario Research Institute, The Ottawa Hospital and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Bruno Eymard
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France ,grid.411439.a0000 0001 2150 9058AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Hanns Lochmüller
- grid.28046.380000 0001 2182 2255Division of Neurology, Department of Medicine, Children’s Hospital of Eastern Ontario Research Institute, The Ottawa Hospital and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Cécile Martinat
- grid.503216.30000 0004 0618 2124INSERM/UEPS UMR 861, Paris Saclay Université, I-STEM, 91100 Corbeil-Essonnes, France
| | - Laurent Schaeffer
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| |
Collapse
|
28
|
Miyana K, Ishiyama A, Saito Y, Nishino I. Tulobuterol is a potential therapeutic drug in congenital myasthenic syndrome. Pediatr Int 2022; 64:e15115. [PMID: 35396808 DOI: 10.1111/ped.15115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/07/2021] [Accepted: 12/27/2021] [Indexed: 01/05/2023]
Affiliation(s)
- Kaori Miyana
- Department of Pediatrics, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Akihiko Ishiyama
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoshihiko Saito
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
29
|
Donkervoort S, Krause N, Dergai M, Yun P, Koliwer J, Gorokhova S, Geist Hauserman J, Cummings BB, Hu Y, Smith R, Uapinyoying P, Ganesh VS, Ghosh PS, Monaghan KG, Edassery SL, Ferle PE, Silverstein S, Chao KR, Snyder M, Ellingwood S, Bharucha‐Goebel D, Iannaccone ST, Dal Peraro M, Foley AR, Savas JN, Bolduc V, Fasshauer D, Bönnemann CG, Schwake M. BET1 variants establish impaired vesicular transport as a cause for muscular dystrophy with epilepsy. EMBO Mol Med 2021; 13:e13787. [PMID: 34779586 PMCID: PMC8649873 DOI: 10.15252/emmm.202013787] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/26/2022] Open
Abstract
BET1 is required, together with its SNARE complex partners GOSR2, SEC22b, and Syntaxin-5 for fusion of endoplasmic reticulum-derived vesicles with the ER-Golgi intermediate compartment (ERGIC) and the cis-Golgi. Here, we report three individuals, from two families, with severe congenital muscular dystrophy (CMD) and biallelic variants in BET1 (P1 p.(Asp68His)/p.(Ala45Valfs*2); P2 and P3 homozygous p.(Ile51Ser)). Due to aberrant splicing and frameshifting, the variants in P1 result in low BET1 protein levels and impaired ER-to-Golgi transport. Since in silico modeling suggested that p.(Ile51Ser) interferes with binding to interaction partners other than SNARE complex subunits, we set off and identified novel BET1 interaction partners with low affinity for p.(Ile51Ser) BET1 protein compared to wild-type, among them ERGIC-53. The BET1/ERGIC-53 interaction was validated by endogenous co-immunoprecipitation with both proteins colocalizing to the ERGIC compartment. Mislocalization of ERGIC-53 was observed in P1 and P2's derived fibroblasts; while in the p.(Ile51Ser) P2 fibroblasts specifically, mutant BET1 was also mislocalized along with ERGIC-53. Thus, we establish BET1 as a novel CMD/epilepsy gene and confirm the emerging role of ER/Golgi SNAREs in CMD.
Collapse
Affiliation(s)
- Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | - Niklas Krause
- Biochemistry III/Faculty of ChemistryBielefeld UniversityBielefeldGermany
| | - Mykola Dergai
- Department of Fundamental NeurosciencesUniversity of LausanneLausanneSwitzerland
| | - Pomi Yun
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | - Judith Koliwer
- Biochemistry III/Faculty of ChemistryBielefeld UniversityBielefeldGermany
| | - Svetlana Gorokhova
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
- Service de Génétique MédicaleHôpital de la Timone, APHMMarseilleFrance
- INSERM, U1251‐MMGAix‐Marseille UniversitéMarseilleFrance
| | - Janelle Geist Hauserman
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | - Beryl B Cummings
- Center for Mendelian GenomicsProgram in Medical and Population GeneticsBroad Institute of MIT and HarvardCambridgeMAUSA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | | | - Prech Uapinyoying
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
- Research for Genetic MedicineChildren's National Medical CenterWashingtonDCUSA
| | - Vijay S Ganesh
- Center for Mendelian GenomicsProgram in Medical and Population GeneticsBroad Institute of MIT and HarvardCambridgeMAUSA
- Department of NeurologyBrigham & Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Partha S Ghosh
- Department of NeurologyBoston Children's HospitalBostonMAUSA
| | | | - Seby L Edassery
- Department of NeurologyFeinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Pia E Ferle
- Biochemistry III/Faculty of ChemistryBielefeld UniversityBielefeldGermany
| | - Sarah Silverstein
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
- Rutgers New Jersey School of MedicineNewarkNJUSA
- Undiagnosed Diseases ProgramNational Human Genome Research InstituteNational Institute of HealthBethesdaMDUSA
| | - Katherine R Chao
- Center for Mendelian GenomicsProgram in Medical and Population GeneticsBroad Institute of MIT and HarvardCambridgeMAUSA
| | - Molly Snyder
- Department of NeurologyChildren's HealthDallasTXUSA
| | | | - Diana Bharucha‐Goebel
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
- Division of NeurologyChildren’s National Medical CenterWashingtonDCUSA
| | - Susan T Iannaccone
- Division of Pediatric NeurologyDepartments of Pediatrics, Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Matteo Dal Peraro
- Institute of BioengineeringSchool of Life SciencesÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | - Jeffrey N Savas
- Department of NeurologyFeinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | - Dirk Fasshauer
- Department of Fundamental NeurosciencesUniversity of LausanneLausanneSwitzerland
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMDUSA
| | - Michael Schwake
- Biochemistry III/Faculty of ChemistryBielefeld UniversityBielefeldGermany
- Department of NeurologyFeinberg School of MedicineNorthwestern UniversityChicagoILUSA
| |
Collapse
|
30
|
Estephan EP, Zambon AA, Thompson R, Polavarapu K, Jomaa D, Töpf A, Helito PVP, Heise CO, Moreno CAM, Silva AMS, Kouyoumdjian JA, Morita MDP, Reed UC, Lochmüller H, Zanoteli E. Congenital myasthenic syndrome: Correlation between clinical features and molecular diagnosis. Eur J Neurol 2021; 29:833-842. [PMID: 34749429 DOI: 10.1111/ene.15173] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To present phenotype features of a large cohort of congenital myasthenic syndromes (CMS) and correlate them with their molecular diagnosis. METHODS Suspected CMS patients were divided into three groups: group A (limb, bulbar or axial weakness, with or without ocular impairment, and all the following: clinical fatigability, electrophysiology compatible with neuromuscular junction involvement and anticholinesterase agents response), group B (limb, bulbar or axial weakness, with or without ocular impairment, and at least one of additional characteristics noted in group A) and group C (pure ocular syndrome). Individual clinical findings and the clinical groups were compared between the group with a confirmed molecular diagnosis of CMS and the group without molecular diagnosis or with a non-CMS molecular diagnosis. RESULTS Seventy-nine patients (68 families) were included in the cohort: 48 in group A, 23 in group B and 8 in group C. Fifty-one were considered confirmed CMS (30 CHRNE, 5 RAPSN, 4 COL13A1, 3 DOK7, 3 COLQ, 2 GFPT1, 1 CHAT, 1 SCN4A, 1 GMPPB, 1 CHRNA1), 7 probable CMS, 5 non-CMS and 16 unsolved. The chance of a confirmed molecular diagnosis of CMS was significantly higher for group A and lower for group C. Some individual clinical features, alterations on biopsy and electrophysiology enhanced specificity for CMS. Muscle imaging showed at least mild alterations in the majority of confirmed cases, with preferential involvement of soleus, especially in CHRNE CMS. CONCLUSIONS Stricter clinical criteria increase the chance of confirming a CMS diagnosis, but may lose sensitivity, especially for some specific genes.
Collapse
Affiliation(s)
- Eduardo P Estephan
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil.,Department of Neurology, Hospital Santa Marcelina, Sao Paulo, Brazil.,Department of Medical Clinic, Faculdade de Medicina Santa Marcelina (FASM), Sao Paulo, Brazil
| | - Antonio A Zambon
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil
| | - Rachel Thompson
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Kiran Polavarapu
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Danny Jomaa
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.,Queen's University School of Medicine, Kingston, ON, Canada
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, University of Newcastle, Newcastle upon Tyne, UK
| | - Paulo V P Helito
- Department of Radiology, Hospital das Clínicas (HCFMUSP), Instituto de Ortopedia (IOT), Sao Paulo, Brazil
| | - Carlos O Heise
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil
| | - Cristiane A M Moreno
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil.,Department of Neurology, Hospital Santa Marcelina, Sao Paulo, Brazil.,Department of Medical Clinic, Faculdade de Medicina Santa Marcelina (FASM), Sao Paulo, Brazil
| | - André M S Silva
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil
| | - Joao A Kouyoumdjian
- Faculdade Estadual de Medicina de Sao Jose do Rio Preto (FAMERP), Sao Jose do Rio Preto, Brazil
| | - Maria da Penha Morita
- Faculdade Estadual de Medicina de Sao Jose do Rio Preto (FAMERP), Sao Jose do Rio Preto, Brazil
| | - Umbertina C Reed
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Medicine, Ottawa Research Institute, Ottawa, ON, Canada.,Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil
| |
Collapse
|
31
|
Sewry CA. Historical aspects of muscle research in the Dubowitz Neuromuscular Centre: the Hammersmith days. Neuromuscul Disord 2021; 31:925-930. [PMID: 34736633 DOI: 10.1016/j.nmd.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Caroline A Sewry
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health 30 Guildford Street, London, WC1N 1EH, United Kingdom; Department of Musculoskeletal Histopathology and the Wolfson Centre for Inherited Neuromuscular Diseases, RJAH Orthopaedic Hospital NHS Trust, Oswestry, SY10 7AG United Kingdom; Department of Cellular Pathology, Salford Royal Hospital NHS Foundation Trust, Northern Care Alliance NHS Group, Stott Lane, Salford M6 8HD United Kingdom.
| |
Collapse
|
32
|
Ramdas S, Beeson D. Congenital myasthenic syndromes: where do we go from here? Neuromuscul Disord 2021; 31:943-954. [PMID: 34736634 DOI: 10.1016/j.nmd.2021.07.400] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 11/27/2022]
Abstract
Congenital myasthenia syndromes are rare but often treatable conditions affecting neuromuscular transmission. They result from loss or impaired function of one of a number of proteins secondary to a genetic defect. An estimate of the prevalence in the UK gave 9.2 cases per million, however, this is likely an underestimate since the adoption of next generation sequencing for diagnosis away from specialist centres is enhancing the 'pick up' rate. Next generation sequencing has helped identify a series of novel genes that harbour mutations causative for congenital myasthenic syndrome that include not only genes that encode proteins specifically expressed at the neuromuscular junction but also those that are ubiquitously expressed. The list of genes harbouring disease-causing mutations for congenital myasthenic syndrome continues to expand and is now over 30, but with many of the newly identified genes it is increasingly being recognised that abnormal neuromuscular transmission is only one component of a multifaceted phenotype in which muscle, the central nervous system, and other organs may also be affected. Treatment can be tailored to the underlying molecular mechanism for impaired neuromuscular transmission but treating the more complex multifaceted disorders and will require development of new therapies.
Collapse
Affiliation(s)
- Sithara Ramdas
- MDUK Neuromuscular centre, Children's Hospital, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - David Beeson
- Neurosciences Group, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford OX3 9DS, UK.
| |
Collapse
|
33
|
Gilhus NE, Verschuuren JJGM, Hovland SIB, Simmonds H, Groot F, Palace J. Myasthenia gravis: do not forget the patient perspective. Neuromuscul Disord 2021; 31:S0960-8966(21)00583-6. [PMID: 34635387 DOI: 10.1016/j.nmd.2021.07.396] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Nils Erik Gilhus
- Department of Clinical Medicine, University of Bergen, Norway; Department of Neurology, Haukeland University Hospital, Bergen, Norway.
| | | | | | - Huw Simmonds
- Myaware, College Business Centre, Derby, England
| | - Floor Groot
- Dutch Neuromuscular disease Association, Baarn, The Netherlands
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Hospitals Trust, Oxford, England
| |
Collapse
|
34
|
Webster RG, Vanhaesebrouck AE, Maxwell SE, Cossins JA, Liu W, Ueta R, Yamanashi Y, Beeson DMW. Effect of salbutamol on neuromuscular junction function and structure in a mouse model of DOK7 congenital myasthenia. Hum Mol Genet 2021; 29:2325-2336. [PMID: 32543656 PMCID: PMC7424765 DOI: 10.1093/hmg/ddaa116] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022] Open
Abstract
Congenital myasthenic syndromes (CMS) are characterized by fatigable muscle weakness resulting from impaired neuromuscular transmission. β2-adrenergic agonists are an effective treatment for DOK7-CMS. DOK7 is a component within the AGRN-LRP4-MUSK-DOK7 signalling pathway that is key for the formation and maintenance of the synaptic structure of the neuromuscular junction (NMJ). The precise mechanism of action of β2-adrenergic agonists at the NMJ is not fully understood. In this study, we investigated whether β2-adrenergic agonists improve both neurotransmission and structural integrity of the NMJ in a mouse model of DOK7-CMS. Ex-vivo electrophysiological techniques and microscopy of the NMJ were used to study the effect of salbutamol, a β2-adrenergic agonist, on synaptic structure and function. DOK7-CMS model mice displayed a severe phenotype with reduced weight gain and perinatal lethality. Salbutamol treatment improved weight gain and survival in DOK7 myasthenic mice. Model animals had fewer active NMJs, detectable by endplate recordings, compared with age-matched wild-type littermates. Salbutamol treatment increased the number of detectable NMJs during endplate recording. Correspondingly, model mice had fewer acetylcholine receptor-stained NMJs detected by fluorescent labelling, but following salbutamol treatment an increased number were detectable. The data demonstrate that salbutamol can prolong survival and increase NMJ number in a severe model of DOK7-CMS.
Collapse
Affiliation(s)
- Richard G Webster
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - An E Vanhaesebrouck
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Susan E Maxwell
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Judith A Cossins
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Weiwei Liu
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Ryo Ueta
- Division of Genetics, Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, Tokyo 135-8550, Japan
| | - Yuji Yamanashi
- Division of Genetics, Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, Tokyo 135-8550, Japan
| | - David M W Beeson
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
35
|
Bukharaeva E, Khuzakhmetova V, Dmitrieva S, Tsentsevitsky A. Adrenoceptors Modulate Cholinergic Synaptic Transmission at the Neuromuscular Junction. Int J Mol Sci 2021; 22:ijms22094611. [PMID: 33924758 PMCID: PMC8124642 DOI: 10.3390/ijms22094611] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Adrenoceptor activators and blockers are widely used clinically for the treatment of cardiovascular and pulmonary disorders. More recently, adrenergic agents have also been used to treat neurodegenerative diseases. Recent studies indicate a location of sympathetic varicosities in close proximity to neuromuscular junctions. The pressing question is whether there could be any effects of endo- or exogenous catecholamines on cholinergic neuromuscular transmission. It was shown that the pharmacological stimulation of adrenoceptors, as well as sympathectomy, can affect both acetylcholine release from motor nerve terminals and the functioning of postsynaptic acetylcholine receptors. In this review, we discuss the recent data regarding the effects of adrenergic drugs on neurotransmission at the neuromuscular junction. The elucidation of the molecular mechanisms by which the clinically relevant adrenomimetics and adrenoblockers regulate quantal acetylcholine release from the presynaptic nerve terminals and postsynaptic sensitivity may help in the design of highly effective and well-tolerated sympathomimetics for treating a number of neurodegenerative diseases accompanied by synaptic defects.
Collapse
|
36
|
Yubero D, Natera-de Benito D, Pijuan J, Armstrong J, Martorell L, Fernàndez G, Maynou J, Jou C, Roldan M, Ortez C, Nascimento A, Hoenicka J, Palau F. The Increasing Impact of Translational Research in the Molecular Diagnostics of Neuromuscular Diseases. Int J Mol Sci 2021; 22:4274. [PMID: 33924139 PMCID: PMC8074304 DOI: 10.3390/ijms22084274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
The diagnosis of neuromuscular diseases (NMDs) has been progressively evolving from the grouping of clinical symptoms and signs towards the molecular definition. Optimal clinical, biochemical, electrophysiological, electrophysiological, and histopathological characterization is very helpful to achieve molecular diagnosis, which is essential for establishing prognosis, treatment and genetic counselling. Currently, the genetic approach includes both the gene-targeted analysis in specific clinically recognizable diseases, as well as genomic analysis based on next-generation sequencing, analyzing either the clinical exome/genome or the whole exome or genome. However, as of today, there are still many patients in whom the causative genetic variant cannot be definitely established and variants of uncertain significance are often found. In this review, we address these drawbacks by incorporating two additional biological omics approaches into the molecular diagnostic process of NMDs. First, functional genomics by introducing experimental cell and molecular biology to analyze and validate the variant for its biological effect in an in-house translational diagnostic program, and second, incorporating a multi-omics approach including RNA-seq, metabolomics, and proteomics in the molecular diagnosis of neuromuscular disease. Both translational diagnostics programs and omics are being implemented as part of the diagnostic process in academic centers and referral hospitals and, therefore, an increase in the proportion of neuromuscular patients with a molecular diagnosis is expected. This improvement in the process and diagnostic performance of patients will allow solving aspects of their health problems in a precise way and will allow them and their families to take a step forward in their lives.
Collapse
Affiliation(s)
- Dèlia Yubero
- Department of Genetic and Molecular Medicine—IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.Y.); (J.A.); (L.M.); (G.F.); (J.M.); (M.R.)
- Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, 08950 Barcelona, Spain;
| | - Daniel Natera-de Benito
- Neuromuscular Unit, Department of Pediatric Neurology, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.N.-d.B.); (C.O.)
| | - Jordi Pijuan
- Laboratory of Neurogenetics and Molecular Medicine—IPER, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Judith Armstrong
- Department of Genetic and Molecular Medicine—IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.Y.); (J.A.); (L.M.); (G.F.); (J.M.); (M.R.)
- Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, 08950 Barcelona, Spain;
| | - Loreto Martorell
- Department of Genetic and Molecular Medicine—IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.Y.); (J.A.); (L.M.); (G.F.); (J.M.); (M.R.)
- Laboratory of Neurogenetics and Molecular Medicine—IPER, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Guerau Fernàndez
- Department of Genetic and Molecular Medicine—IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.Y.); (J.A.); (L.M.); (G.F.); (J.M.); (M.R.)
- Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, 08950 Barcelona, Spain;
| | - Joan Maynou
- Department of Genetic and Molecular Medicine—IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.Y.); (J.A.); (L.M.); (G.F.); (J.M.); (M.R.)
- Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, 08950 Barcelona, Spain;
| | - Cristina Jou
- Department of Pathology, Hospital Sant Joan de Déu, Pediatric Biobank for Research, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Mònica Roldan
- Department of Genetic and Molecular Medicine—IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.Y.); (J.A.); (L.M.); (G.F.); (J.M.); (M.R.)
- Confocal Microscopy and Cellular Imaging Unit, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Carlos Ortez
- Neuromuscular Unit, Department of Pediatric Neurology, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.N.-d.B.); (C.O.)
- Division of Pediatrics, Clinic Institute of Medicine & Dermatology, Hospital Clínic, University of Barcelona School of Medicine and Health Sciences, 08950 Barcelona, Spain
| | - Andrés Nascimento
- Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, 08950 Barcelona, Spain;
- Neuromuscular Unit, Department of Pediatric Neurology, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.N.-d.B.); (C.O.)
| | - Janet Hoenicka
- Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, 08950 Barcelona, Spain;
- Laboratory of Neurogenetics and Molecular Medicine—IPER, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Francesc Palau
- Department of Genetic and Molecular Medicine—IPER, Hospital Sant Joan de Déu and Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (D.Y.); (J.A.); (L.M.); (G.F.); (J.M.); (M.R.)
- Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, 08950 Barcelona, Spain;
- Laboratory of Neurogenetics and Molecular Medicine—IPER, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
- Department of Pathology, Hospital Sant Joan de Déu, Pediatric Biobank for Research, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain;
| |
Collapse
|
37
|
Díaz-Santiago E, Claros MG, Yahyaoui R, de Diego-Otero Y, Calvo R, Hoenicka J, Palau F, Ranea JAG, Perkins JR. Decoding Neuromuscular Disorders Using Phenotypic Clusters Obtained From Co-Occurrence Networks. Front Mol Biosci 2021; 8:635074. [PMID: 34046427 PMCID: PMC8147726 DOI: 10.3389/fmolb.2021.635074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Neuromuscular disorders (NMDs) represent an important subset of rare diseases associated with elevated morbidity and mortality whose diagnosis can take years. Here we present a novel approach using systems biology to produce functionally-coherent phenotype clusters that provide insight into the cellular functions and phenotypic patterns underlying NMDs, using the Human Phenotype Ontology as a common framework. Gene and phenotype information was obtained for 424 NMDs in OMIM and 126 NMDs in Orphanet, and 335 and 216 phenotypes were identified as typical for NMDs, respectively. ‘Elevated serum creatine kinase’ was the most specific to NMDs, in agreement with the clinical test of elevated serum creatinine kinase that is conducted on NMD patients. The approach to obtain co-occurring NMD phenotypes was validated based on co-mention in PubMed abstracts. A total of 231 (OMIM) and 150 (Orphanet) clusters of highly connected co-occurrent NMD phenotypes were obtained. In parallel, a tripartite network based on phenotypes, diseases and genes was used to associate NMD phenotypes with functions, an approach also validated by literature co-mention, with KEGG pathways showing proportionally higher overlap than Gene Ontology and Reactome. Phenotype-function pairs were crossed with the co-occurrent NMD phenotype clusters to obtain 40 (OMIM) and 72 (Orphanet) functionally coherent phenotype clusters. As expected, many of these overlapped with known diseases and confirmed existing knowledge. Other clusters revealed interesting new findings, indicating informative phenotypes for differential diagnosis, providing deeper knowledge of NMDs, and pointing towards specific cell dysfunction caused by pleiotropic genes. This work is an example of reproducible research that i) can help better understand NMDs and support their diagnosis by providing a new tool that exploits existing information to obtain novel clusters of functionally-related phenotypes, and ii) takes us another step towards personalised medicine for NMDs.
Collapse
Affiliation(s)
- Elena Díaz-Santiago
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, Málaga, Spain
| | - M Gonzalo Claros
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, Málaga, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain.,Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, Málaga, Spain.,Institute for Mediterranean and Subtropical Horticulture "La Mayora" (IHSM-UMA-CSIC), Málaga, Spain
| | - Raquel Yahyaoui
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, Málaga, Spain.,Laboratory of Metabolopathies and Neonatal Screening, Málaga Regional University Hospital, Málaga, Spain
| | | | - Rocío Calvo
- Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, Málaga, Spain.,Laboratory of Metabolopathies and Neonatal Screening, Málaga Regional University Hospital, Málaga, Spain
| | - Janet Hoenicka
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain.,Sant Joan de Déu Hospital and Research Institute, Barcelona, Spain
| | - Francesc Palau
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain.,Sant Joan de Déu Hospital and Research Institute, Barcelona, Spain.,Hospital Clínic and University of Barcelona School of Medicine and Health Sciences, Barcelona, Spain
| | - Juan A G Ranea
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, Málaga, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain.,Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, Málaga, Spain
| | - James R Perkins
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, Málaga, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain.,Institute of Biomedical Research in Malaga (IBIMA), IBIMA-RARE, Málaga, Spain
| |
Collapse
|
38
|
Zhao Y, Li Y, Bian Y, Yao S, Liu P, Yu M, Zhang W, Wang Z, Yuan Y. Congenital myasthenic syndrome in China: genetic and myopathological characterization. Ann Clin Transl Neurol 2021; 8:898-907. [PMID: 33756069 PMCID: PMC8045908 DOI: 10.1002/acn3.51346] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE We aimed to summarize the clinical, genetic, and myopathological features of a cohort of Chinese patients with congenital myasthenic syndrome, and follow up on therapeutic outcomes. METHODS The clinical spectrum, mutational frequency of genes, and pathological diagnostic clues of various subtypes of patients with congenital myasthenic syndrome were summarized. Therapeutic effects were followed up. RESULTS Thirty-five patients from 29 families were recruited. Ten genes were identified: GFPT1 (27.6%), AGRN (17.2%), CHRNE (17.2%), COLQ (13.8%), GMPPB (6.9%), CHAT, CHRNA1, DOK7, COG7, and SLC25A1 (3.4% each, respectively). Sole limb-girdle weakness was found in patients with AGRN (1/8) and GFPT1 (7/8) mutations, whereas distal weakness was all observed in patients with AGRN (6/8) mutations. Tubular aggregates were only found in patients with GFPT1 mutations (5/6). The patients with GMPPB mutations (2/2) had decreased alpha-dystroglycan. Acetylcholinesterase inhibitor therapy resulted in no response or worsened symptoms in patients with COLQ mutations, a diverse response in patients with AGRN mutations, and a good response in patients with other subtypes. Albuterol therapy was effective or harmless in most subtypes. Therapy effects became attenuated with long-term use in patients with COLQ or AGRN mutations. INTERPRETATION The genetic distribution of congenital myasthenic syndrome in China is distinct from that of other ethnic origins. The appearance of distal weakness, selective limb-girdle myasthenic syndrome, tubular aggregates, and decreased alpha-dystroglycan were indicative of the specific subtypes. Based on the follow-up findings, we suggest cautious evaluation of the long-term efficacy of therapeutic agents in congenital myasthenic syndrome.
Collapse
Affiliation(s)
- Yawen Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ying Li
- Department of Neurology, Capital Medical University Affiliated Anzhen Hospital, Chaoyang-qu, China
| | - Yang Bian
- Department of Neurology, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Sheng Yao
- Department of Neurology, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Penju Liu
- Department of Neurology, Capital Medical University Affiliated Anzhen Hospital, Chaoyang-qu, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
39
|
Fernandes M, Caetano A, Pinto M, Medeiros E, Santos L. Diagnosis of DOK7 congenital myasthenic syndrome during pregnancy: A case report and literature review. Clin Neurol Neurosurg 2021; 203:106591. [PMID: 33714798 DOI: 10.1016/j.clineuro.2021.106591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/14/2021] [Accepted: 02/27/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Pregnancy among patients with congenital myasthenic syndrome (CMS) is a rare occurrence. Since most of the patients with CMS reach adulthood, questions regarding clinical outcome with pregnancy arise. CASE REPORT We describe a 38-year-old Portuguese female who presented in the second trimester of pregnancy with proximal fluctuating limb-girdle weakness, hyperlordosis, waddling gait, dysphagia, dysphonia and ptosis, with no ophthalmoparesis. Initial diagnosis of seronegative myasthenia, supported by neurophysiology findings, led to unsuccessful treatment with intravenous immunoglobulin, pyridostigmine, prednisolone and plasmapheresis, and the patient slowly progressed to a severe tetraparesis with facial and bulbar involvement. Genetic testing for CMS identified a novel compound heterozygous mutation (c.1124_1127dupTGCC and c.935_936del) in the DOK7 gene. Subsequent treatment with salbutamol resulted in substantial clinical benefit. CONCLUSIONS This case underlines the importance of considering the diagnosis of CMS in patients with fluctuating weakness during pregnancy. Patients of child-bearing potential diagnosed with CMS, particularly due to DOK7 mutations, should be counseled in advance and closely followed during pregnancy.
Collapse
Affiliation(s)
- Marco Fernandes
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126, 1349-019, Lisbon, Portugal.
| | - André Caetano
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126, 1349-019, Lisbon, Portugal; CEDOC Chronic Diseases Research Centre, Nova Medical School / Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Miguel Pinto
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126, 1349-019, Lisbon, Portugal
| | - Elmira Medeiros
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126, 1349-019, Lisbon, Portugal; CEDOC Chronic Diseases Research Centre, Nova Medical School / Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Luís Santos
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126, 1349-019, Lisbon, Portugal; CEDOC Chronic Diseases Research Centre, Nova Medical School / Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
40
|
Paz ML, Barrantes FJ. Cholesterol in myasthenia gravis. Arch Biochem Biophys 2021; 701:108788. [PMID: 33548213 DOI: 10.1016/j.abb.2021.108788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/13/2021] [Accepted: 01/26/2021] [Indexed: 01/03/2023]
Abstract
The cholinergic neuromuscular junction is the paradigm peripheral synapse between a motor neuron nerve ending and a skeletal muscle fiber. In vertebrates, acetylcholine is released from the presynaptic site and binds to the nicotinic acetylcholine receptor at the postsynaptic membrane. A variety of pathologies among which myasthenia gravis stands out can impact on this rapid and efficient signaling mechanism, including autoimmune diseases affecting the nicotinic receptor or other synaptic proteins. Cholesterol is an essential component of biomembranes and is particularly rich at the postsynaptic membrane, where it interacts with and modulates many properties of the nicotinic receptor. The profound changes inflicted by myasthenia gravis on the postsynaptic membrane necessarily involve cholesterol. This review analyzes some aspects of myasthenia gravis pathophysiology and associated postsynaptic membrane dysfunction, including dysregulation of cholesterol metabolism in the myocyte brought about by antibody-receptor interactions. In addition, given the extensive therapeutic use of statins as the typical cholesterol-lowering drugs, we discuss their effects on skeletal muscle and the possible implications for MG patients under chronic treatment with this type of compound.
Collapse
Affiliation(s)
- Mariela L Paz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra de Inmunología, Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral "Prof. Dr. Ricardo A. Margni" (IDEHU), Buenos Aires, Argentina
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute (BIOMED), UCA, CONICET, Av. Alicia Moreau de Justo 1600, C1107AFF, Buenos Aires, Argentina.
| |
Collapse
|
41
|
Lamond A, Buckley D, O'Dea J, Turner L. Variants of SLC18A3 leading to congenital myasthenic syndrome in two children with varying presentations. BMJ Case Rep 2021; 14:14/1/e237799. [PMID: 33462016 PMCID: PMC7813295 DOI: 10.1136/bcr-2020-237799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
This report describes the variation in presentation of two unrelated patients found to have a rare form of presynaptic congenital myasthenic syndrome. Both patients presented with hypotonia, ptosis, poor weight gain and apneic episodes. Through whole exome sequencing, our patients were found to have the same likely pathogenic biallelic variants in W315X and I200N of SLC18A3, encoding vesicular acetylcholine transporter (VAChT). These specific variants in SLC18A3 have not been previously described in the literature. We illustrate the variety in clinical presentation and course of children with mutations in SLC18A3, leading to presynaptic congenital myasthenic syndrome through VAChT deficiency.
Collapse
Affiliation(s)
- Allison Lamond
- Pediatrics, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| | - David Buckley
- Pediatric Neurology, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| | - Jennifer O'Dea
- Pediatrics, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| | - Lesley Turner
- Genetics, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
42
|
Rodríguez Cruz PM, Hughes I, Manzur A, Munot P, Ramdas S, Wright R, Breen C, Pitt M, Pagnamenta AT, Taylor JC, Palace J, Beeson D. Presynaptic congenital myasthenic syndrome due to three novel mutations in SLC5A7 encoding the sodium-dependant high-affinity choline transporter. Neuromuscul Disord 2021; 31:21-28. [DOI: 10.1016/j.nmd.2020.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 01/14/2023]
|
43
|
Bhat S, El-Kasaby A, Freissmuth M, Sucic S. Functional and Biochemical Consequences of Disease Variants in Neurotransmitter Transporters: A Special Emphasis on Folding and Trafficking Deficits. Pharmacol Ther 2020; 222:107785. [PMID: 33310157 PMCID: PMC7612411 DOI: 10.1016/j.pharmthera.2020.107785] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023]
Abstract
Neurotransmitters, such as γ-aminobutyric acid, glutamate, acetyl choline, glycine and the monoamines, facilitate the crosstalk within the central nervous system. The designated neurotransmitter transporters (NTTs) both release and take up neurotransmitters to and from the synaptic cleft. NTT dysfunction can lead to severe pathophysiological consequences, e.g. epilepsy, intellectual disability, or Parkinson’s disease. Genetic point mutations in NTTs have recently been associated with the onset of various neurological disorders. Some of these mutations trigger folding defects in the NTT proteins. Correct folding is a prerequisite for the export of NTTs from the endoplasmic reticulum (ER) and the subsequent trafficking to their pertinent site of action, typically at the plasma membrane. Recent studies have uncovered some of the key features in the molecular machinery responsible for transporter protein folding, e.g., the role of heat shock proteins in fine-tuning the ER quality control mechanisms in cells. The therapeutic significance of understanding these events is apparent from the rising number of reports, which directly link different pathological conditions to NTT misfolding. For instance, folding-deficient variants of the human transporters for dopamine or GABA lead to infantile parkinsonism/dystonia and epilepsy, respectively. From a therapeutic point of view, some folding-deficient NTTs are amenable to functional rescue by small molecules, known as chemical and pharmacological chaperones.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
44
|
Bauché S, Sureau A, Sternberg D, Rendu J, Buon C, Messéant J, Boëx M, Furling D, Fauré J, Latypova X, Gelot AB, Mayer M, Mary P, Whalen S, Fournier E, Cloix I, Remerand G, Laffargue F, Nougues MC, Fontaine B, Eymard B, Isapof A, Strochlic L. New recessive mutations in SYT2 causing severe presynaptic congenital myasthenic syndromes. NEUROLOGY-GENETICS 2020; 6:e534. [PMID: 33659639 PMCID: PMC7803339 DOI: 10.1212/nxg.0000000000000534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/25/2020] [Indexed: 11/15/2022]
Abstract
Objective To report the identification of 2 new homozygous recessive mutations in the synaptotagmin 2 (SYT2) gene as the genetic cause of severe and early presynaptic forms of congenital myasthenic syndromes (CMSs). Methods Next-generation sequencing identified new homozygous intronic and frameshift mutations in the SYT2 gene as a likely cause of presynaptic CMS. We describe the clinical and electromyographic patient phenotypes, perform ex vivo splicing analyses to characterize the effect of the intronic mutation on exon splicing, and analyze the functional impact of this variation at the neuromuscular junction (NMJ). Results The 2 infants presented a similar clinical phenotype evoking first a congenital myopathy characterized by muscle weakness and hypotonia. Next-generation sequencing allowed to the identification of 1 homozygous intronic mutation c.465+1G>A in patient 1 and another homozygous frameshift mutation c.328_331dup in patient 2, located respectively in the 5' splice donor site of SYT2 intron 4 and in exon 3. Functional studies of the intronic mutation validated the abolition of the splice donor site of exon 4 leading to its skipping. In-frame skipping of exon 4 that encodes part of the C2A calcium-binding domain of SYT2 is associated with a loss-of-function effect resulting in a decrease of neurotransmitter release and severe pre- and postsynaptic NMJ defects. Conclusions This study identifies new homozygous recessive SYT2 mutations as the underlying cause of severe and early presynaptic form of CMS expanding the genetic spectrum of recessive SYT2-related CMS associated with defects in neurotransmitter release.
Collapse
Affiliation(s)
- Stéphanie Bauché
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Alain Sureau
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Damien Sternberg
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - John Rendu
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Céline Buon
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Julien Messéant
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Myriam Boëx
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Denis Furling
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Julien Fauré
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Xénia Latypova
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Antoinette Bernabe Gelot
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Michèle Mayer
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Pierre Mary
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Sandra Whalen
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Emmanuel Fournier
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Isabelle Cloix
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Ganaelle Remerand
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Fanny Laffargue
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Marie-Christine Nougues
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Bertrand Fontaine
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Bruno Eymard
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Arnaud Isapof
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Laure Strochlic
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| |
Collapse
|
45
|
Rodríguez Cruz PM, Cossins J, Beeson D, Vincent A. The Neuromuscular Junction in Health and Disease: Molecular Mechanisms Governing Synaptic Formation and Homeostasis. Front Mol Neurosci 2020; 13:610964. [PMID: 33343299 PMCID: PMC7744297 DOI: 10.3389/fnmol.2020.610964] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/30/2020] [Indexed: 12/28/2022] Open
Abstract
The neuromuscular junction (NMJ) is a highly specialized synapse between a motor neuron nerve terminal and its muscle fiber that are responsible for converting electrical impulses generated by the motor neuron into electrical activity in the muscle fibers. On arrival of the motor nerve action potential, calcium enters the presynaptic terminal, which leads to the release of the neurotransmitter acetylcholine (ACh). ACh crosses the synaptic gap and binds to ACh receptors (AChRs) tightly clustered on the surface of the muscle fiber; this leads to the endplate potential which initiates the muscle action potential that results in muscle contraction. This is a simplified version of the events in neuromuscular transmission that take place within milliseconds, and are dependent on a tiny but highly structured NMJ. Much of this review is devoted to describing in more detail the development, maturation, maintenance and regeneration of the NMJ, but first we describe briefly the most important molecules involved and the conditions that affect their numbers and function. Most important clinically worldwide, are myasthenia gravis (MG), the Lambert-Eaton myasthenic syndrome (LEMS) and congenital myasthenic syndromes (CMS), each of which causes specific molecular defects. In addition, we mention the neurotoxins from bacteria, snakes and many other species that interfere with neuromuscular transmission and cause potentially fatal diseases, but have also provided useful probes for investigating neuromuscular transmission. There are also changes in NMJ structure and function in motor neuron disease, spinal muscle atrophy and sarcopenia that are likely to be secondary but might provide treatment targets. The NMJ is one of the best studied and most disease-prone synapses in the nervous system and it is amenable to in vivo and ex vivo investigation and to systemic therapies that can help restore normal function.
Collapse
Affiliation(s)
- Pedro M. Rodríguez Cruz
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Judith Cossins
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
46
|
Qiao R, Di F, Wang J, Wei Y, Zhang Y, Xu T, Wang Y, Gu W, Han B, Yang R. The Association Between RAPSN Methylation in Peripheral Blood and Early Stage Lung Cancer Detected in Case-Control Cohort. Cancer Manag Res 2020; 12:11063-11075. [PMID: 33173339 PMCID: PMC7646459 DOI: 10.2147/cmar.s275321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/03/2020] [Indexed: 12/24/2022] Open
Abstract
Background Early detection is essential to improve the survival and life quality of lung cancer (LC) patients. Changes of peripheral blood DNA methylation could be associated with malignancy but were mostly studied in Caucasians. Methods Here, in a Chinese population, we performed mass spectrometry assays to investigate the association between very early stage LC and methylation levels of RAPSN in the peripheral blood by a case–control cohort using of 221 LC patients (93.2% LC at stage I) and 285 unrelated cancer free control individuals. Results The odds ratios (ORs) of all CpG sites were evaluated for their risk to LC using inter-quartile analyses by logistic regression. In general, we observed an association between very early LC and decreased methylation of RAPSN_CpG_1.15 and RAPSN_CpG_3.4 (referring to Q4, OR range from 1.64 to 1.81, p<0.05). Stratified by gender, while hypomethylation of RAPSN_CpG_1.15, RAPSN_CpG_3.4 and RAPSN_CpG_7.14 were associated with LC in males (referring to Q4, ORs range from 1.94 to 2.31, p<0.05), RAPSN_CpG_2 and RAPSN_CpG_5 showed significantly lower methylation in female LC patients comparing to controls (referring to Q4, ORs range from 2.49 to 3.60, p<0.05). The risk of RAPSN hypomethylation to LC was enhanced by aging, and typically for people older than 55 years (referring to Q4, ORs range from 2.17 to 3.61 in six out of all 10 analyzed CpG groups, p<0.05). Conclusion Our study reveals an association between RAPSN hypomethylation in peripheral blood and LC and suggests the occurrence of altered blood-based methylation at the early stage of cancer.
Collapse
Affiliation(s)
- Rong Qiao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, People's Republic of China
| | - Feifei Di
- Department of Research and Academic, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing 210000, People's Republic of China
| | - Jun Wang
- Department of Research and Academic, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing 210000, People's Republic of China
| | - Yujie Wei
- Department of Research and Academic, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing 210000, People's Republic of China
| | - Yanman Zhang
- Department of Research and Academic, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing 210000, People's Republic of China
| | - Tian Xu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210000, People's Republic of China
| | - Yue Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, People's Republic of China
| | - Wanjian Gu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210000, People's Republic of China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, People's Republic of China
| | - Rongxi Yang
- Department of Research and Academic, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing 210000, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210000, People's Republic of China
| |
Collapse
|
47
|
Caldas VM, Heise CO, Kouyoumdjian JA, Zambon AA, Silva AMS, Estephan EDP, Zanoteli E. Electrophysiological study of neuromuscular junction in congenital myasthenic syndromes, congenital myopathies, and chronic progressive external ophthalmoplegia. Neuromuscul Disord 2020; 30:897-903. [PMID: 33121830 DOI: 10.1016/j.nmd.2020.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/19/2022]
Abstract
This study was designed to analyze the sensitivity, specificity, and accuracy of jitter parameters combined with repetitive nerve stimulation (RNS) in congenital myasthenic syndrome (CMS), chronic progressive external ophthalmoplegia (CPEO), and congenital myopathies (CM). Jitter was obtained with a concentric needle electrode during voluntary activation of the Orbicularis Oculi muscle in CMS (n = 21), CPEO (n = 20), and CM (n = 18) patients and in controls (n = 14). RNS (3 Hz) was performed in six different muscles for all patients (Abductor Digiti Minimi, Tibialis Anterior, upper Trapezius, Deltoideus, Orbicularis Oculi, and Nasalis). RNS was abnormal in 90.5% of CMS patients and in only one CM patient. Jitter was abnormal in 95.2% of CMS, 20% of CPEO, and 11.1% of CM patients. No patient with CPEO or CM presented a mean jitter higher than 53.6 µs or more than 30% abnormal individual jitter (> 45 µs). No patient with CPEO or CM and mild abnormal jitter values presented an abnormal decrement. Jitter and RNS assessment are valuable tools for diagnosing neuromuscular transmission abnormalities in CMS patients. A mean jitter value above 53.6 µs or the presence of more than 30% abnormal individual jitter (> 45 µs) strongly suggests CMS compared with CPEO and CM.
Collapse
Affiliation(s)
- Vitor Marques Caldas
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Av. Dr. Enéas Carvalho Aguiar 255, 05403-900 São Paulo SP, Brazil
| | - Carlos Otto Heise
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Av. Dr. Enéas Carvalho Aguiar 255, 05403-900 São Paulo SP, Brazil
| | - João Aris Kouyoumdjian
- Laboratório de Investigação Neuromuscular, Faculdade Estadual de Medicina de São Jose do Rio Preto (FAMERP), São Jose do Rio Preto, SP, Brazil
| | - Antônio Alberto Zambon
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Av. Dr. Enéas Carvalho Aguiar 255, 05403-900 São Paulo SP, Brazil
| | - André Macedo Serafim Silva
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Av. Dr. Enéas Carvalho Aguiar 255, 05403-900 São Paulo SP, Brazil
| | - Eduardo de Paula Estephan
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Av. Dr. Enéas Carvalho Aguiar 255, 05403-900 São Paulo SP, Brazil; Department of Medicine, Faculdade Santa Marcelina, São Paulo, Brazil
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Av. Dr. Enéas Carvalho Aguiar 255, 05403-900 São Paulo SP, Brazil.
| |
Collapse
|
48
|
Cossins J, Webster R, Maxwell S, Rodríguez Cruz PM, Knight R, Llewelyn JG, Shin JY, Palace J, Beeson D. Congenital myasthenic syndrome due to a TOR1AIP1 mutation: a new disease pathway for impaired synaptic transmission. Brain Commun 2020; 2:fcaa174. [PMID: 33215087 PMCID: PMC7660151 DOI: 10.1093/braincomms/fcaa174] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/24/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
Congenital myasthenic syndromes are inherited disorders characterized by fatiguable muscle weakness resulting from impaired signal transmission at the neuromuscular junction. Causative mutations have been identified in genes that can affect the synaptic function or structure. We identified a homozygous frameshift deletion c.127delC, p. Pro43fs in TOR1AIP1 in two siblings with limb-girdle weakness and impaired transmission at the neuromuscular synapse. TOR1AIP1 encodes the inner nuclear membrane protein lamin-associated protein 1. On muscle biopsy from the index case, lamin-associated protein 1 was absent from myonuclei. A mouse model with lamin-associated protein 1 conditionally knocked out in striated muscle was used to analyse the role of lamin-associated protein 1 in synaptic dysfunction. Model mice develop fatiguable muscle weakness as demonstrated by using an inverted screen hang test. Electromyography on the mice revealed a decrement on repetitive nerve stimulation. Ex vivo analysis of hemi-diaphragm preparations showed both miniature and evoked end-plate potential half-widths were prolonged which was associated with upregulation of the foetal acetylcholine receptor γ subunit. Neuromuscular junctions on extensor digitorum longus muscles were enlarged and fragmented, and the number of subsynaptic nuclei was significantly increased. Following these findings, electromyography was performed on cases of other nuclear envelopathies caused by mutations in LaminA/C or emerin, but decrement on repetitive nerve stimulation or other indications of defective neuromuscular transmission were not seen. Thus, this report highlights the first nuclear membrane protein in which defective function can lead to impaired synaptic transmission.
Collapse
Affiliation(s)
- Judith Cossins
- Neurosciences Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Richard Webster
- Neurosciences Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Susan Maxwell
- Neurosciences Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Pedro M Rodríguez Cruz
- Neurosciences Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Ravi Knight
- Department of Clinical Neurology, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - John Gareth Llewelyn
- Neurology Department, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Ji-Yeon Shin
- Department of Medicine, Columbia University Medical Centre, New York, NY 10032, USA
| | - Jacqueline Palace
- Department of Clinical Neurology, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - David Beeson
- Neurosciences Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
49
|
Tang BL. SNAREs and developmental disorders. J Cell Physiol 2020; 236:2482-2504. [PMID: 32959907 DOI: 10.1002/jcp.30067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Members of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family mediate membrane fusion processes associated with vesicular trafficking and autophagy. SNAREs mediate core membrane fusion processes essential for all cells, but some SNAREs serve cell/tissue type-specific exocytic/endocytic functions, and are therefore critical for various aspects of embryonic development. Mutations or variants of their encoding genes could give rise to developmental disorders, such as those affecting the nervous system and immune system in humans. Mutations to components in the canonical synaptic vesicle fusion SNARE complex (VAMP2, STX1A/B, and SNAP25) and a key regulator of SNARE complex formation MUNC18-1, produce variant phenotypes of autism, intellectual disability, movement disorders, and epilepsy. STX11 and MUNC18-2 mutations underlie 2 subtypes of familial hemophagocytic lymphohistiocytosis. STX3 mutations contribute to variant microvillus inclusion disease. Chromosomal microdeletions involving STX16 play a role in pseudohypoparathyroidism type IB associated with abnormal imprinting of the GNAS complex locus. In this short review, I discuss these and other SNARE gene mutations and variants that are known to be associated with a variety developmental disorders, with a focus on their underlying cellular and molecular pathological basis deciphered through disease modeling. Possible pathogenic potentials of other SNAREs whose variants could be disease predisposing are also speculated upon.
Collapse
Affiliation(s)
- Bor L Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
50
|
Cetin H, Beeson D, Vincent A, Webster R. The Structure, Function, and Physiology of the Fetal and Adult Acetylcholine Receptor in Muscle. Front Mol Neurosci 2020; 13:581097. [PMID: 33013323 PMCID: PMC7506097 DOI: 10.3389/fnmol.2020.581097] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/13/2020] [Indexed: 12/31/2022] Open
Abstract
The neuromuscular junction (NMJ) is a highly developed synapse linking motor neuron activity with muscle contraction. A complex of molecular cascades together with the specialized NMJ architecture ensures that each action potential arriving at the motor nerve terminal is translated into an action potential in the muscle fiber. The muscle-type nicotinic acetylcholine receptor (AChR) is a key molecular component located at the postsynaptic muscle membrane responsible for the generation of the endplate potential (EPP), which usually exceeds the threshold potential necessary to activate voltage-gated sodium channels and triggers a muscle action potential. Two AChR isoforms are found in mammalian muscle. The fetal isoform is present in prenatal stages and is involved in the development of the neuromuscular system whereas the adult isoform prevails thereafter, except after denervation when the fetal form is re-expressed throughout the muscle. This review will summarize the structural and functional differences between the two isoforms and outline congenital and autoimmune myasthenic syndromes that involve the isoform specific AChR subunits.
Collapse
Affiliation(s)
- Hakan Cetin
- Department of Neurology, Medical University of Vienna, Vienna, Austria.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Richard Webster
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|