1
|
Kosmanopoulos GP, Donohue JK, Hoke M, Thomas S, Peyton MA, Vo L, Crawford TO, Sadjadi R, Herrmann DN, Yum SW, Reilly MM, Scherer SS, Finkel RS, Lewis RA, Pareyson D, Pisciotta C, Walk D, Shy ME, Sumner CJ, McCray BA. TRPV4 neuromuscular disease registry highlights bulbar, skeletal and proximal limb manifestations. Brain 2025; 148:238-251. [PMID: 38917025 PMCID: PMC12054732 DOI: 10.1093/brain/awae201] [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: 04/04/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Dominant missense mutations of the calcium-permeable cation channel TRPV4 cause Charcot-Marie-Tooth disease (CMT) type 2C and two forms of distal spinal muscular atrophy. These conditions are collectively referred to as TRPV4-related neuromuscular disease and share features of motor greater than sensory dysfunction and frequent vocal fold weakness. Pathogenic variants lead to gain of ion channel function that can be rescued by TRPV4 antagonists in cellular and animal models. As small molecule TRPV4 antagonists have proven safe in trials for other disease indications, channel inhibition is a promising therapeutic strategy for TRPV4 patients. However, the current knowledge of the clinical features and natural history of TRPV4-related neuromuscular disease is insufficient to enable rational clinical trial design. To address these issues, we developed a TRPV4 patient database and administered a TRPV4-specific patient questionnaire. Here, we report demographic and clinical information, including CMT Examination Scores (CMTES), from 68 patients with known pathogenic TRPV4 variants, 40 of whom also completed the TRPV4 patient questionnaire. TRPV4 patients showed a bimodal age of onset, with the largest peak occurring in the first 2 years of life. Compared to CMT type 1A (CMT1A) patients, TRPV4 patients showed distinct symptoms and signs, manifesting more ambulatory difficulties and more frequent involvement of proximal arm and leg muscles. Although patients reported fewer sensory symptoms, sensory dysfunction was often detected clinically. Many patients were affected by vocal fold weakness (55%) and shortness of breath (55%), and 11% required ventilatory support. Skeletal abnormalities were common, including scoliosis (64%), arthrogryposis (33%) and foot deformities. Strikingly, patients with infantile onset of disease showed less sensory involvement and less progression of symptoms. These results highlight distinctive clinical features in TRPV4 patients, including motor-predominant disease, proximal arm and leg weakness, severe ambulatory difficulties, vocal fold weakness, respiratory dysfunction and skeletal involvement. In addition, patients with infantile onset of disease appeared to have a distinct phenotype with less apparent disease progression based on CMTES. These collective observations indicate that clinical trial design for TRPV4-related neuromuscular disease should include outcome measures that reliably capture non-length dependent motor dysfunction, vocal fold weakness and respiratory disease.
Collapse
Affiliation(s)
- Gage P Kosmanopoulos
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jack K Donohue
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Maya Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Simone Thomas
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Margo A Peyton
- Department of Neurology, Mass General Brigham, Boston, MA 02114, USA
| | - Linh Vo
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Thomas O Crawford
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Reza Sadjadi
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David N Herrmann
- Department of Neurology, University of Rochester, Rochester, NY 14627, USA
| | - Sabrina W Yum
- Department of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mary M Reilly
- Centre for Neuromuscular Diseases, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Steven S Scherer
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Richard S Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richard A Lewis
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Davide Pareyson
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Chiara Pisciotta
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - David Walk
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brett A McCray
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
2
|
Liu M, Duan Y, Dong J, Zhang K, Jin X, Gao M, Jia H, Chen J, Liu M, Wei M, Zhong X. Early signs of neurodegenerative diseases: Possible mechanisms and targets for Golgi stress. Biomed Pharmacother 2024; 175:116646. [PMID: 38692058 DOI: 10.1016/j.biopha.2024.116646] [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: 02/28/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
The Golgi apparatus plays a crucial role in mediating the modification, transport, and sorting of intracellular proteins and lipids. The morphological changes occurring in the Golgi apparatus are exceptionally important for maintaining its function. When exposed to external pressure or environmental stimulation, the Golgi apparatus undergoes adaptive changes in both structure and function, which are known as Golgi stress. Although certain signal pathway responses or post-translational modifications have been observed following Golgi stress, further research is needed to comprehensively summarize and understand the related mechanisms. Currently, there is evidence linking Golgi stress to neurodegenerative diseases; however, the role of Golgi stress in the progression of neurodegenerative diseases such as Alzheimer's disease remains largely unexplored. This review focuses on the structural and functional alterations of the Golgi apparatus during stress, elucidating potential mechanisms underlying the involvement of Golgi stress in regulating immunity, autophagy, and metabolic processes. Additionally, it highlights the pivotal role of Golgi stress as an early signaling event implicated in the pathogenesis and progression of neurodegenerative diseases. Furthermore, this study summarizes prospective targets that can be therapeutically exploited to mitigate neurodegenerative diseases by targeting Golgi stress. These findings provide a theoretical foundation for identifying novel breakthroughs in preventing and treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Mengyu Liu
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Ying Duan
- Liaoning Maternal and Child Health Hospital, Shayang, Liaoning 110005, China
| | - Jianru Dong
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Kaisong Zhang
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Menglin Gao
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Huachao Jia
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Ju Chen
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Mingyan Liu
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China.
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang, Liaoning 110167, China.
| | - Xin Zhong
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
3
|
Fernández-Eulate G, Theuriet J, Record CJ, Querin G, Masingue M, Leonard-Louis S, Behin A, Le Forestier N, Pegat A, Michaud M, Chanson JB, Nadaj-Pakleza A, Tard C, Bedat-Millet AL, Sole G, Spinazzi M, Salort-Campana E, Echaniz-Laguna A, Poinsignon V, Latour P, Reilly MM, Bouhour F, Stojkovic T. Phenotype Presentation and Molecular Diagnostic Yield in Non-5q Spinal Muscular Atrophy. Neurol Genet 2023; 9:e200087. [PMID: 37470033 PMCID: PMC10352921 DOI: 10.1212/nxg.0000000000200087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/26/2023] [Indexed: 07/21/2023]
Abstract
Background and Objectives Spinal muscular atrophy (SMA) is mainly caused by homozygous SMN1 gene deletions on 5q13. Non-5q SMA patients' series are lacking, and the diagnostic yield of next-generation sequencing (NGS) is largely unknown. The aim of this study was to describe the clinical and genetic landscape of non-5q SMA and evaluate the performance of neuropathy gene panels in these disorders. Methods Description of patients with non-5q SMA followed in the different neuromuscular reference centers in France as well as in London, United Kingdom. Patients without a genetic diagnosis had undergone at least a neuropathy or large neuromuscular gene panel. Results Seventy-one patients from 65 different families were included, mostly sporadic cases (60.6%). At presentation, 21 patients (29.6%) showed exclusive proximal weakness (P-SMA), 35 (49.3%) showed associated distal weakness (PD-SMA), and 15 (21.1%) a scapuloperoneal phenotype (SP-SMA). Thirty-two patients (45.1%) had a genetic diagnosis: BICD2 (n = 9), DYNC1H1 (n = 7), TRPV4 (n = 4), VCP, HSBP1, AR (n = 2), VRK1, DNAJB2, MORC2, ASAH1, HEXB, and unexpectedly, COL6A3 (n = 1). The genetic diagnostic yield was lowest in P-SMA (6/21, 28.6%) compared with PD-SMA (16/35, 45.7%) and SP-SMA (10/15, 66.7%). An earlier disease onset and a family history of the disease or consanguinity were independent predictors of a positive genetic diagnosis. Neuropathy gene panels were performed in 59 patients with a 32.2% diagnostic yield (19/59). In 13 additional patients, a genetic diagnosis was achieved through individual gene sequencing or an alternative neuromuscular NGS. Discussion Non-5q SMA is genetically heterogeneous, and neuropathy gene panels achieve a molecular diagnosis in one-third of the patients. The diagnostic yield can be increased by sequencing of other neuromuscular and neurometabolic genes. Nevertheless, there is an unmet need to cluster these patients to aid in the identification of new genes.
Collapse
Affiliation(s)
- Gorka Fernández-Eulate
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Julian Theuriet
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Christopher J Record
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Giorgia Querin
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Marion Masingue
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Sarah Leonard-Louis
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Anthony Behin
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Nadine Le Forestier
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Antoine Pegat
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Maud Michaud
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Jean-Baptiste Chanson
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Aleksandra Nadaj-Pakleza
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Celine Tard
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Anne-Laure Bedat-Millet
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Guilhem Sole
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Marco Spinazzi
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Emmanuelle Salort-Campana
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Andoni Echaniz-Laguna
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Vianney Poinsignon
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Philippe Latour
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Mary M Reilly
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Francoise Bouhour
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| | - Tanya Stojkovic
- From the Nord/Est/Ile-de-France Neuromuscular Reference Center (G.F.-E., G.Q., M. Masingue, S.L.-L., A.B., T.S.), Institut de Myologie, Pitié-Salpêtrière Hospital, Paris; Electromyography and Neuromuscular Department (J.T., A.P., F.B.), Hospices Civils de Lyon; Centre for Neuromuscular Diseases (C.J.R., M.M.R.), UCL Queen Square Institute of Neurology, London, United Kingdom; Neurology Department (N.L.F.), Pitié-Salpêtrière Hospital, Paris; Nord/Est/Ile-de-France Neuromuscular Reference Center (M. Michaud), Central Nancy University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (J.-B.C., A.N.-P.), Strasbourg University Hospitals; Nord/Est/Ile-de-France Neuromuscular Reference Center (C.T.), U1172, Lille University Hospital; Nord/Est/Ile-de-France Neuromuscular Reference Center (A.-L.B.-M.), Rouen University Hospital; Neuromuscular Reference Center 'AOC' (G.S.), Bordeaux University Hospitals (Pellegrin Hospital); Neuromuscular Reference Center (M.S.), Angers University Hospital; Neuromuscular and ALS Reference Center (E.S.-C.), La Timone University Hospital, Marseille; French National Center for Rare Neuropathies (A.E.-L.), Neurology Department, Bicêtre University Hospital, INSERM U1195, Paris-Saclay University; Molecular Genetics Lab (V.P.), Bicêtre University Hospital, Le Kremlin Bicêtre; and Center for Biology - East (P.L.), Neurological Hereditary Disorders Unit, Hospices Civils de Lyon, France
| |
Collapse
|
4
|
Lazo PA, Morejón-García P. VRK1 variants at the cross road of Cajal body neuropathogenic mechanisms in distal neuropathies and motor neuron diseases. Neurobiol Dis 2023; 183:106172. [PMID: 37257665 DOI: 10.1016/j.nbd.2023.106172] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023] Open
Abstract
Distal hereditary neuropathies and neuro motor diseases are complex neurological phenotypes associated with pathogenic variants in a large number of genes, but in some the origin is unknown. Recently, rare pathogenic variants of the human VRK1 gene have been associated with these neurological phenotypes. All VRK1 pathogenic variants are recessive, and their clinical presentation occurs in either homozygous or compound heterozygous patients. The pathogenic VRK1 gene pathogenic variants are located in three clusters within the protein sequence. The main, and initial, shared clinical phenotype among VRK1 pathogenic variants is a distal progressive loss of motor and/or sensory function, which includes diseases such as spinal muscular atrophy, Charcot-Marie-Tooth, amyotrophic lateral sclerosis and hereditary spastic paraplegia. In most cases, symptoms start early in infancy, or in utero, and are slowly progressive. Additional neurological symptoms vary among non-related patients, probably because of their different VRK1 variants and their genetic background. The underlying common pathogenic mechanism, by its functional impairment, is a likely consequence of the roles that the VRK1 protein plays in the regulation on the stability and assembly of Cajal bodies, which affect RNA maturation and processing, neuronal migration of RNPs along axons, and DNA-damage responses. Alterations of these processes are associated with several neuro sensory or motor syndromes. The clinical heterogeneity of the neurological phenotypes associated with VRK1 is a likely consequence of the protein complexes in which VRK1 is integrated, which include several proteins known to be associated with Cajal bodies and DNA damage responses. Several hereditary distal neurological diseases are a consequence of pathogenic variants in genes that alter these cellular functions. We conclude that VRK1-related distal hereditary neuropathies and motor neuron diseases represent a novel subgroup of Cajal body related neurological syndromes.
Collapse
Affiliation(s)
- Pedro A Lazo
- Molecular Mechanisms of Cancer Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain.
| | - Patricia Morejón-García
- Molecular Mechanisms of Cancer Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain.
| |
Collapse
|
5
|
Pinto WBVDR, Souza PVSD, Badia BML, Farias IB, Albuquerque Filho JMVD, Gonçalves EA, Machado RIL, Oliveira ASB. Adult-onset non-5q proximal spinal muscular atrophy: a comprehensive review. ARQUIVOS DE NEURO-PSIQUIATRIA 2021; 79:912-923. [PMID: 34706022 DOI: 10.1590/0004-282x-anp-2020-0429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/24/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND Adult-onset spinal muscular atrophy (SMA) represents an expanding group of inherited neurodegenerative disorders in clinical practice. OBJECTIVE This review aims to synthesize the main clinical, genetic, radiological, biochemical, and neurophysiological aspects related to the classical and recently described forms of proximal SMA. METHODS The authors performed a non-systematic critical review summarizing adult-onset proximal SMA presentations. RESULTS Previously limited to cases of SMN1-related SMA type 4 (adult form), this group has now more than 15 different clinical conditions that have in common the symmetrical and progressive compromise of lower motor neurons starting in adulthood or elderly stage. New clinical and genetic subtypes of adult-onset proximal SMA have been recognized and are currently target of wide neuroradiological, pathological, and genetic studies. CONCLUSIONS This new complex group of rare disorders typically present with lower motor neuron disease in association with other neurological or systemic signs of impairment, which are relatively specific and typical for each genetic subtype.
Collapse
Affiliation(s)
| | - Paulo Victor Sgobbi de Souza
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | - Bruno Mattos Lombardi Badia
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | - Igor Braga Farias
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | | | - Eduardo Augusto Gonçalves
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | - Roberta Ismael Lacerda Machado
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | - Acary Souza Bulle Oliveira
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| |
Collapse
|
6
|
Abstract
RNA-binding proteins are a critical group of multifunctional proteins that precisely regulate all aspects of gene expression, from alternative splicing to mRNA trafficking, stability, and translation. Converging evidence highlights aberrant RNA metabolism as a common pathogenic mechanism in several neurodevelopmental and neurodegenerative diseases. However, dysregulation of disease-linked RNA-binding proteins results in widespread, often tissue-specific and/or pleiotropic effects on the transcriptome, making it challenging to determine the underlying cellular and molecular mechanisms that contribute to disease pathogenesis. Understanding how splicing misregulation as well as alterations of mRNA stability and localization impact the activity and function of neuronal proteins is fundamental to addressing neurodevelopmental defects and synaptic dysfunction in disease. Here we highlight recent exciting studies that use high-throughput transcriptomic analysis and advanced genetic, cell biological, and imaging approaches to dissect the role of disease-linked RNA-binding proteins on different RNA processing steps. We focus specifically on efforts to elucidate the functional consequences of aberrant RNA processing on neuronal morphology, synaptic activity and plasticity in development and disease. We also consider new areas of investigation that will elucidate the molecular mechanisms RNA-binding proteins use to achieve spatiotemporal control of gene expression for neuronal homeostasis and plasticity.
Collapse
Affiliation(s)
- Shavanie Prashad
- Department of Pathology, Yale University School of Medicine, Yale University, New Haven, CT, USA.,Experimental Pathology Graduate Group, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Pallavi P Gopal
- Department of Pathology, Yale University School of Medicine, Yale University, New Haven, CT, USA.,Experimental Pathology Graduate Group, Yale University School of Medicine, Yale University, New Haven, CT, USA.,Yale Center for RNA Science and Medicine, Yale University School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
7
|
Falgairolle M, O'Donovan MJ. Motoneuronal Spinal Circuits in Degenerative Motoneuron Disease. Front Mol Neurosci 2020; 13:74. [PMID: 32523513 PMCID: PMC7261878 DOI: 10.3389/fnmol.2020.00074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/15/2020] [Indexed: 12/22/2022] Open
Abstract
The most evident phenotype of degenerative motoneuron disease is the loss of motor function which accompanies motoneuron death. In both amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), it is now clear that dysfunction is not restricted to motoneurons but is manifest in the spinal circuits in which motoneurons are embedded. As mounting evidence shows that motoneurons possess more elaborate and extensive connections within the spinal cord than previously realized, it is necessary to consider the role of this circuitry and its dysfunction in the disease process. In this review article, we ask if the selective vulnerability of the different motoneuron types and the relative disease resistance of distinct motoneuron groups can be understood in terms of their intraspinal connections.
Collapse
Affiliation(s)
- Mélanie Falgairolle
- Section on Developmental Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Michael J O'Donovan
- Section on Developmental Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
8
|
Marcos AT, Martín‐Doncel E, Morejón‐García P, Marcos‐Alcalde I, Gómez‐Puertas P, Segura‐Puimedon M, Armengol L, Navarro‐Pando JM, Lazo PA. VRK1 (Y213H) homozygous mutant impairs Cajal bodies in a hereditary case of distal motor neuropathy. Ann Clin Transl Neurol 2020; 7:808-818. [PMID: 32365420 PMCID: PMC7261760 DOI: 10.1002/acn3.51050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Distal motor neuropathies with a genetic origin have a heterogeneous clinical presentation with overlapping features affecting distal nerves and including spinal muscular atrophies and amyotrophic lateral sclerosis. This indicates that their genetic background is heterogeneous. PATIENT AND METHODS In this work, we have identified and characterized the genetic and molecular base of a patient with a distal sensorimotor neuropathy of unknown origin. For this study, we performed whole-exome sequencing, molecular modelling, cloning and expression of mutant gene, and biochemical and cell biology analysis of the mutant protein. RESULTS A novel homozygous recessive mutation in the human VRK1 gene, coding for a chromatin kinase, causing a substitution (c.637T > C; p.Tyr213His) in exon 8, was detected in a patient presenting since childhood a progressive distal sensorimotor neuropathy and spinal muscular atrophy syndrome, with normal intellectual development. Molecular modelling predicted this mutant VRK1 has altered the kinase activation loop by disrupting its interaction with the C-terminal regulatory region. The p.Y213H mutant protein has a reduced kinase activity with different substrates, including histones H3 and H2AX, proteins involved in DNA damage responses, such as p53 and 53BP1, and coilin, the scaffold for Cajal bodies. The mutant VRK1(Y213H) protein is unable to rescue the formation of Cajal bodies assembled on coilin, in the absence of wild-type VRK1. CONCLUSION The VRK1(Y213H) mutant protein alters the activation loop, impairs the kinase activity of VRK1 causing a functional insufficiency that impairs the formation of Cajal bodies assembled on coilin, a protein that regulates SMN1 and Cajal body formation.
Collapse
Affiliation(s)
- Ana T. Marcos
- Unidad de GenéticaInstituto para el Estudio de la Biología de la Reproducción Humana (INEBIR)SevillaSpain
| | - Elena Martín‐Doncel
- Molecular Mechanisms of Cancer ProgramInstituto de Biología Molecular y Celular del CáncerConsejo Superior de Investigaciones Científicas (CSIC)Universidad de SalamancaSalamancaSpain
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de SalamancaSalamancaSpain
| | - Patricia Morejón‐García
- Molecular Mechanisms of Cancer ProgramInstituto de Biología Molecular y Celular del CáncerConsejo Superior de Investigaciones Científicas (CSIC)Universidad de SalamancaSalamancaSpain
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de SalamancaSalamancaSpain
| | - Iñigo Marcos‐Alcalde
- Molecular Modelling GroupCentro de Biología Molecular “Severo Ochoa”CSIC‐Universidad Autónoma de Madrid, CantoblancoMadridSpain
- School of Experimental SciencesBiosciences Research InstituteUniversidad Francisco de VitoriaPozuelo de Alarcón, MadridSpain
| | - Paulino Gómez‐Puertas
- Molecular Modelling GroupCentro de Biología Molecular “Severo Ochoa”CSIC‐Universidad Autónoma de Madrid, CantoblancoMadridSpain
| | - María Segura‐Puimedon
- Quantitative Genomic Medicine Laboratories, qGenomicsEspluges de LlobregatBarcelonaSpain
| | - Lluis Armengol
- Quantitative Genomic Medicine Laboratories, qGenomicsEspluges de LlobregatBarcelonaSpain
| | - José M. Navarro‐Pando
- Unidad de GenéticaInstituto para el Estudio de la Biología de la Reproducción Humana (INEBIR)SevillaSpain
- Cátedra de Reproducción y Genética HumanaFacultad de Ciencias de la SaludUniversidad Europea del AtlánticoSantanderSpain
- Fundación Universitaria Iberoamericana (FUNIBER)BarcelonaSpain
| | - Pedro A. Lazo
- Molecular Mechanisms of Cancer ProgramInstituto de Biología Molecular y Celular del CáncerConsejo Superior de Investigaciones Científicas (CSIC)Universidad de SalamancaSalamancaSpain
- Instituto de Investigación Biomédica de Salamanca (IBSAL)Hospital Universitario de SalamancaSalamancaSpain
| |
Collapse
|
9
|
Sedghi M, Moslemi AR, Olive M, Etemadifar M, Ansari B, Nasiri J, Emrahi L, Mianesaz HR, Laing NG, Tajsharghi H. Motor neuron diseases caused by a novel VRK1 variant - A genotype/phenotype study. Ann Clin Transl Neurol 2019; 6:2197-2204. [PMID: 31560180 PMCID: PMC6856620 DOI: 10.1002/acn3.50912] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/16/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background Motor neuron disorders involving upper and lower neurons are a genetically and clinically heterogenous group of rare neuromuscular disorders with overlap among spinal muscular atrophies (SMAs) and amyotrophic lateral sclerosis (ALS). Classical SMA caused by recessive mutations in SMN1 is one of the most common genetic causes of mortality in infants. It is characterized by degeneration of anterior horn cells in the spinal cord, leading to progressive muscle weakness and atrophy. Non‐SMN1‐related spinal muscular atrophies are caused by variants in a number of genes, including VRK1, encoding the vaccinia‐related kinase 1 (VRK1). VRK1 variants have been segregated with motor neuron diseases including SMA phenotypes or hereditary complex motor and sensory axonal neuropathy (HMSN), with or without pontocerebellar hypoplasia or microcephaly. Results Here, we report an association of a novel homozygous splice variant in VRK1 (c.1159 + 1G>A) with childhood‐onset SMA or juvenile lower motor disease with brisk tendon reflexes without pontocerebellar hypoplasia and normal intellectual ability in a family with five affected individuals. We show that the VRK1 splice variant in patients causes decreased splicing efficiency and a mRNA frameshift that escapes the nonsense‐mediated decay machinery and results in a premature termination codon. Conclusions Our findings unveil the impact of the variant on the VRK1 transcript and further support the implication of VRK1 in the pathogenesis of lower motor neuron diseases.
Collapse
Affiliation(s)
- Maryam Sedghi
- Medical Genetics Laboratory, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali-Reza Moslemi
- Department of Pathology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Montse Olive
- Institute of Neuropathology, Department of Pathology, Institut Investigació Biomèdica de Bellvitge (IDIBELL)-Hospital de Bellvitge, Hospitalet de Llobregat, 08907, Barcelona, Spain.,Neuromuscular Unit, Department of Neurology, Institut Investigació Biomèdica de Bellvitge-(IDIBELL)-Hospital de Bellvitge, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Masoud Etemadifar
- Department of Functional Neursurgery, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behnaz Ansari
- Department of Neurology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jafar Nasiri
- Department of Pediatric Neurology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Emrahi
- Department of Pathology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hamid-Reza Mianesaz
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nigel G Laing
- Centre for Medical Research, The University of Western Australia and the Harry Perkins Institute for Medical Research, Nedlands, Western Australia, Australia
| | - Homa Tajsharghi
- Centre for Medical Research, The University of Western Australia and the Harry Perkins Institute for Medical Research, Nedlands, Western Australia, Australia.,School of Health Sciences, Division Biomedicine and Translational Medicine, University of Skovde, Skovde, Sweden
| |
Collapse
|
10
|
Evaluating Benefit-risk Decision-making in Spinal Muscular Atrophy: A First-ever Study to Assess Risk Tolerance in the SMA Patient Community. Clin Ther 2019; 41:943-960.e4. [PMID: 31056304 DOI: 10.1016/j.clinthera.2019.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/08/2019] [Accepted: 03/21/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE Patients' perceptions of benefit-risk are essential to informing the regulatory process and the context in which potential therapies are evaluated. To bring this critical information to regulators, Cure SMA launched a first-ever Benefit-Risk Survey for spinal muscular atrophy (SMA) to characterize decision-making and benefit-risk trade-offs in SMA associated with a potential therapy. We hypothesized that risk tolerance would be correlated with SMA type/severity and disease progression. This article presents the results of a benefit-risk survey to enhance understanding of how patients with SMA and caregivers evaluate specific benefits and risks associated with potential therapies. METHODS Affected adults, representing all SMA types (I-IV) within the Cure SMA database, and caregivers of affected individuals of all ages/types were invited via e-mail to participate. Best-worst scaling (BWS) was used to assess participants' priorities on benefit-risk trade-offs, as it provides higher discrimination and importance scaling among tested attributes. Twelve potentially clinically meaningful treatment benefits and 11 potential risks (ranging in severity and immediacy) were tested. Multiple factors were correlated with individual responses, including: SMA type/disease severity, stage of disease, respondent type, sex, and quality of life/level of independence (current and expected). Survey respondents were also evaluated for "risk-taking attitudes." FINDINGS A total of 298 responses were evaluated (28% affected adults and 72% caregivers, mostly parents). Most respondents were diagnosed >5 years ago (67.3%), with 22.1% SMA type I, 45.6% SMA type II, and 27.9% SMA type III. No strong correlation was found between risk tolerance and SMA type, stage of disease progression, respondent type, sex, quality of life assessment, or rated levels of independence. Irrespective of SMA type, respondents consistently rated the following risks, associated with a potential treatment, as "least tolerable": life-threatening allergic reactions; 1 in 1000 risk of life-threatening side effects leading to possible organ failure; or worsening quality of life. Furthermore, all SMA type respondents rated these risks as "most tolerable": invasive mode of treatment administration (including need for general anesthesia); side effect of dizziness; and other common side effects such as nausea, vomiting, loss of appetite, headaches, back pain, or fatigue. IMPLICATIONS With the approval of the first SMA treatment, these findings offer a unique opportunity to assess and characterize baseline risk-tolerance in SMA against which to evaluate future SMA treatment options. Although differences had been expected in risk tolerance among respondents based on disease baseline and certain patient attributes, this was not observed. Survey results should inform future SMA drug development and benefit-risk assessments.
Collapse
|
11
|
Karakaya M, Storbeck M, Strathmann EA, Delle Vedove A, Hölker I, Altmueller J, Naghiyeva L, Schmitz-Steinkrüger L, Vezyroglou K, Motameny S, Alawbathani S, Thiele H, Polat AI, Okur D, Boostani R, Karimiani EG, Wunderlich G, Ardicli D, Topaloglu H, Kirschner J, Schrank B, Maroofian R, Magnusson O, Yis U, Nürnberg P, Heller R, Wirth B. Targeted sequencing with expanded gene profile enables high diagnostic yield in non-5q-spinal muscular atrophies. Hum Mutat 2018; 39:1284-1298. [PMID: 29858556 DOI: 10.1002/humu.23560] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/14/2018] [Accepted: 05/30/2018] [Indexed: 11/08/2022]
Abstract
Spinal muscular atrophies (SMAs) are a heterogeneous group of disorders characterized by muscular atrophy, weakness, and hypotonia due to suspected lower motor neuron degeneration (LMND). In a large cohort of 3,465 individuals suspected with SMA submitted for SMN1 testing to our routine diagnostic laboratory, 48.8% carried a homozygous SMN1 deletion, 2.8% a subtle mutation, and an SMN1 deletion, whereas 48.4% remained undiagnosed. Recently, several other genes implicated in SMA/LMND have been reported. Despite several efforts to establish a diagnostic algorithm for non-5q-SMA (SMA without deletion or point mutations in SMN1 [5q13.2]), data from large-scale studies are not available. We tested the clinical utility of targeted sequencing in non-5q-SMA by developing two different gene panels. We first analyzed 30 individuals with a small panel including 62 genes associated with LMND using IonTorrent-AmpliSeq target enrichment. Then, additional 65 individuals were tested with a broader panel encompassing up to 479 genes implicated in neuromuscular diseases (NMDs) with Agilent-SureSelect target enrichment. The NMD panel provided a higher diagnostic yield (33%) than the restricted LMND panel (13%). Nondiagnosed cases were further subjected to exome or genome sequencing. Our experience supports the use of gene panels covering a broad disease spectrum for diseases that are highly heterogeneous and clinically difficult to differentiate.
Collapse
Affiliation(s)
- Mert Karakaya
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Markus Storbeck
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Eike A Strathmann
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Andrea Delle Vedove
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Irmgard Hölker
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Janine Altmueller
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany.,Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Leyla Naghiyeva
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Lea Schmitz-Steinkrüger
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Katharina Vezyroglou
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Susanne Motameny
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Salem Alawbathani
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Holger Thiele
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Ayse Ipek Polat
- Dokuz Eylül University, Department of Pediatric Neurology, Izmir, Turkey
| | - Derya Okur
- Dokuz Eylül University, Department of Pediatric Neurology, Izmir, Turkey
| | - Reza Boostani
- Mashhad University of Medical Sciences, Department of Neurology, Mashhad, Iran
| | - Ehsan Ghayoor Karimiani
- Next Generation Genetic Polyclinic, Mashhad, Iran.,Razavi Cancer Research Center, Razavi Hospital, Imam Reza International University, Mashhad, Iran
| | | | - Didem Ardicli
- Hacettepe University, Department of Pediatric Neurology, Ankara, Turkey
| | - Haluk Topaloglu
- Hacettepe University, Department of Pediatric Neurology, Ankara, Turkey
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Bertold Schrank
- DKD HELIOS Kliniken, Department of Neurology, Wiesbaden, Germany
| | - Reza Maroofian
- Genetics and Molecular Cell Sciences Research Centre, St George's University of London, London, UK
| | | | - Uluc Yis
- Dokuz Eylül University, Department of Pediatric Neurology, Izmir, Turkey
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Raoul Heller
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
12
|
Gray KM, Kaifer KA, Baillat D, Wen Y, Bonacci TR, Ebert AD, Raimer AC, Spring AM, Have ST, Glascock JJ, Gupta K, Van Duyne GD, Emanuele MJ, Lamond AI, Wagner EJ, Lorson CL, Matera AG. Self-oligomerization regulates stability of survival motor neuron protein isoforms by sequestering an SCF Slmb degron. Mol Biol Cell 2018; 29:96-110. [PMID: 29167380 PMCID: PMC5909936 DOI: 10.1091/mbc.e17-11-0627] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022] Open
Abstract
Spinal muscular atrophy (SMA) is caused by homozygous mutations in human SMN1 Expression of a duplicate gene (SMN2) primarily results in skipping of exon 7 and production of an unstable protein isoform, SMNΔ7. Although SMN2 exon skipping is the principal contributor to SMA severity, mechanisms governing stability of survival motor neuron (SMN) isoforms are poorly understood. We used a Drosophila model system and label-free proteomics to identify the SCFSlmb ubiquitin E3 ligase complex as a novel SMN binding partner. SCFSlmb interacts with a phosphor degron embedded within the human and fruitfly SMN YG-box oligomerization domains. Substitution of a conserved serine (S270A) interferes with SCFSlmb binding and stabilizes SMNΔ7. SMA-causing missense mutations that block multimerization of full-length SMN are also stabilized in the degron mutant background. Overexpression of SMNΔ7S270A, but not wild-type (WT) SMNΔ7, provides a protective effect in SMA model mice and human motor neuron cell culture systems. Our findings support a model wherein the degron is exposed when SMN is monomeric and sequestered when SMN forms higher-order multimers.
Collapse
Affiliation(s)
- Kelsey M Gray
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Kevin A Kaifer
- Molecular Pathogenesis and Therapeutics Program, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211
| | - David Baillat
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77550
| | - Ying Wen
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Thomas R Bonacci
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Amanda C Raimer
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Ashlyn M Spring
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| | - Sara Ten Have
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD15EH, UK
| | - Jacqueline J Glascock
- Molecular Pathogenesis and Therapeutics Program, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211
| | - Kushol Gupta
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Gregory D Van Duyne
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Michael J Emanuele
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD15EH, UK
| | - Eric J Wagner
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77550
| | - Christian L Lorson
- Molecular Pathogenesis and Therapeutics Program, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211
| | - A Gregory Matera
- Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
- Integrative Program in Biological and Genome Sciences, Department of Biology and Department of Genetics, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
13
|
Balak CD, Hunter JM, Ahearn ME, Wiley D, D'urso G, Baumbach-Reardon L. Functional characterizations of rare UBA1 variants in X-linked Spinal Muscular Atrophy. F1000Res 2017; 6:1636. [PMID: 29034082 PMCID: PMC5615770 DOI: 10.12688/f1000research.11878.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 02/05/2023] Open
Abstract
Background: X-linked spinal muscular atrophy (XL-SMA) results from mutations in the Ubiquitin-Like Modifier Activating Enzyme 1 (
UBA1). Previously, four novel closely clustered mutations have been shown to cause this fatal infantile disorder affecting only males. These mutations, three missense and one synonymous, all lie within Exon15 of the
UBA1 gene, which contains the active adenylation domain (AAD). Methods: In this study, our group characterized the three known missense variants
in vitro. Using a novel Uba1 assay and other methods, we investigated Uba1 adenylation, thioester, and transthioesterification reactions
in vitro to determine possible biochemical effects of the missense variants. Results: Our data revealed that only one of the three XL-SMA missense variants impairs the Ubiquitin-adenylating ability of Uba1. Additionally, these missense variants retained Ubiquitin thioester bond formation and transthioesterification rates equal to that found in the wild type. Conclusions: Our results demonstrate a surprising shift from the likelihood of these XL-SMA mutations playing a damaging role in Uba1’s enzymatic activity with Ubiquitin, to other roles such as altering
UBA1 mRNA splicing via the disruption of splicing factor binding sites, similar to a mechanism in traditional SMA, or disrupting binding to other important
in vivo binding partners. These findings help to narrow the search for the areas of possible dysfunction in the Ubiquitin-proteasome pathway that ultimately result in XL-SMA. Moreover, this investigation provides additional critical understanding of the mutations’ biochemical mechanisms, vital for the development of future effective diagnostic assays and therapeutics.
Collapse
Affiliation(s)
- Chris D Balak
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, 85004, USA
| | - Jesse M Hunter
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, 85004, USA.,Clinical Genomics, Ambry Genetics, 15 Argonaut, Aliso Viejo, California , 92656, USA
| | - Mary E Ahearn
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, 85004, USA
| | - David Wiley
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida , 33101, USA
| | - Gennaro D'urso
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida , 33101, USA
| | - Lisa Baumbach-Reardon
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, 85004, USA.,Department of Medicine, Division of Clinical Data Analytics and Decision Support, University of Arizona, College of Medicine-Phoenix, Arizona , 85004, USA
| |
Collapse
|
14
|
Prior R, Van Helleputte L, Benoy V, Van Den Bosch L. Defective axonal transport: A common pathological mechanism in inherited and acquired peripheral neuropathies. Neurobiol Dis 2017; 105:300-320. [DOI: 10.1016/j.nbd.2017.02.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/29/2017] [Accepted: 02/20/2017] [Indexed: 12/29/2022] Open
|
15
|
Rietz A, Li H, Quist KM, Cherry JJ, Lorson CL, Burnett BG, Kern NL, Calder AN, Fritsche M, Lusic H, Boaler PJ, Choi S, Xing X, Glicksman MA, Cuny GD, Androphy EJ, Hodgetts KJ. Discovery of a Small Molecule Probe That Post-Translationally Stabilizes the Survival Motor Neuron Protein for the Treatment of Spinal Muscular Atrophy. J Med Chem 2017; 60:4594-4610. [PMID: 28481536 DOI: 10.1021/acs.jmedchem.6b01885] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Spinal muscular atrophy (SMA) is the leading genetic cause of infant death. We previously developed a high-throughput assay that employs an SMN2-luciferase reporter allowing identification of compounds that act transcriptionally, enhance exon recognition, or stabilize the SMN protein. We describe optimization and characterization of an analog suitable for in vivo testing. Initially, we identified analog 4m that had good in vitro properties but low plasma and brain exposure in a mouse PK experiment due to short plasma stability; this was overcome by reversing the amide bond and changing the heterocycle. Thiazole 27 showed excellent in vitro properties and a promising mouse PK profile, making it suitable for in vivo testing. This series post-translationally stabilizes the SMN protein, unrelated to global proteasome or autophagy inhibition, revealing a novel therapeutic mechanism that should complement other modalities for treatment of SMA.
Collapse
Affiliation(s)
- Anne Rietz
- Department of Dermatology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Hongxia Li
- Department of Dermatology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Kevin M Quist
- Department of Dermatology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Jonathan J Cherry
- Department of Dermatology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Christian L Lorson
- Department of Veterinary Pathobiology, Bond Life Sciences Center, University of Missouri , Columbia, Missouri 65201, United States
| | - Barrington G Burnett
- Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland 20814, United States
| | - Nicholas L Kern
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Alyssa N Calder
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Melanie Fritsche
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Hrvoje Lusic
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Patrick J Boaler
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Sungwoon Choi
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Xuechao Xing
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Marcie A Glicksman
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Gregory D Cuny
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Elliot J Androphy
- Department of Dermatology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Kevin J Hodgetts
- Laboratory for Drug Discovery in Neurodegeneration, Brigham & Women's Hospital and Harvard Medical School , 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
16
|
Abstract
OBJECTIVE In the past decade, hereditary forms of motor neuron disease (spinal muscular atrophy and/or amyotrophic lateral sclerosis) are increasingly identified. As advanced genetic testing is performed, molecular diagnosis can be obtained. Identifying new gene mutations can lead to further understanding of disease. METHODS AND RESULTS We report a single case of a patient with early-onset amyotrophic lateral sclerosis, evaluated at University of Texas Health Houston Science Center from 2011-2014. Initial genetic testing did not reveal an etiology in this patient. Through whole-exome sequencing, a VRK1 mutation was identified. CONCLUSIONS AND RELEVANCE We identify a possible new cause of hereditary amyotrophic lateral sclerosis, VRK1 mutation. This case report also expands the phenotypic spectrum of this mutation in neurologic diseases.
Collapse
|
17
|
Martinez-Carrera LA, Wirth B. Dominant spinal muscular atrophy is caused by mutations in BICD2, an important golgin protein. Front Neurosci 2015; 9:401. [PMID: 26594138 PMCID: PMC4633519 DOI: 10.3389/fnins.2015.00401] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/09/2015] [Indexed: 11/19/2022] Open
Abstract
Spinal muscular atrophies (SMAs) are characterized by degeneration of spinal motor neurons and muscle weakness. Autosomal recessive SMA is the most common form and is caused by homozygous deletions/mutations of the SMN1 gene. However, families with dominant inherited SMA have been reported, for most of them the causal gene remains unknown. Recently, we and others have identified heterozygous mutations in BICD2 as causative for autosomal dominant SMA, lower extremity-predominant, 2 (SMALED2) and hereditary spastic paraplegia (HSP). BICD2 encodes the Bicaudal D2 protein, which is considered to be a golgin, due to its coiled-coil (CC) structure and interaction with the small GTPase RAB6A located at the Golgi apparatus. Golgins are resident proteins in the Golgi apparatus and form a matrix that helps to maintain the structure of this organelle. Golgins are also involved in the regulation of vesicle transport. In vitro overexpression experiments and studies of fibroblast cell lines derived from patients, showed fragmentation of the Golgi apparatus. In the current review, we will discuss possible causes for this disruption, and the consequences at cellular level, with a view to better understand the pathomechanism of this disease.
Collapse
Affiliation(s)
- Lilian A Martinez-Carrera
- Institute of Human Genetics, Institute for Genetics and Center for Molecular Medicine of The University of Cologne Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, Institute for Genetics and Center for Molecular Medicine of The University of Cologne Cologne, Germany
| |
Collapse
|
18
|
The spinal muscular atrophy with pontocerebellar hypoplasia gene VRK1 regulates neuronal migration through an amyloid-β precursor protein-dependent mechanism. J Neurosci 2015; 35:936-42. [PMID: 25609612 DOI: 10.1523/jneurosci.1998-14.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Spinal muscular atrophy with pontocerebellar hypoplasia (SMA-PCH) is an infantile SMA variant with additional manifestations, particularly severe microcephaly. We previously identified a nonsense mutation in Vaccinia-related kinase 1 (VRK1), R358X, as a cause of SMA-PCH. VRK1-R358X is a rare founder mutation in Ashkenazi Jews, and additional mutations in patients of different origins have recently been identified. VRK1 is a nuclear serine/threonine protein kinase known to play multiple roles in cellular proliferation, cell cycle regulation, and carcinogenesis. However, VRK1 was not known to have neuronal functions before its identification as a gene mutated in SMA-PCH. Here we show that VRK1-R358X homozygosity results in lack of VRK1 protein, and demonstrate a role for VRK1 in neuronal migration and neuronal stem cell proliferation. Using shRNA in utero electroporation in mice, we show that Vrk1 knockdown significantly impairs cortical neuronal migration, and affects the cell cycle of neuronal progenitors. Expression of wild-type human VRK1 rescues both proliferation and migration phenotypes. However, kinase-dead human VRK1 rescues only the migration impairment, suggesting the role of VRK1 in neuronal migration is partly noncatalytic. Furthermore, we found that VRK1 deficiency in human and mouse leads to downregulation of amyloid-β precursor protein (APP), a known neuronal migration gene. APP overexpression rescues the phenotype caused by Vrk1 knockdown, suggesting that VRK1 affects neuronal migration through an APP-dependent mechanism.
Collapse
|
19
|
Generation of neurospheres from human adipose-derived stem cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:743714. [PMID: 25815334 PMCID: PMC4357140 DOI: 10.1155/2015/743714] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/24/2015] [Accepted: 02/10/2015] [Indexed: 01/29/2023]
Abstract
Transplantation of neural stem cells (NSCs) to treat neurodegenerative disease shows promise; however, the clinical application of NSCs is limited by the invasive procurement and ethical concerns. Adipose-derived stem cells (ADSCs) are a source of multipotent stem cells that can self-renew and differentiate into various kinds of cells; this study intends to generate neurospheres from human ADSCs by culturing ADSCs on uncoated culture flasks in serum-free neurobasal medium supplemented with B27, basic fibroblast growth factor (bFGF), and epidermal growth factor (EGF); the ADSCs-derived neurospheres were terminally differentiated after growth factor withdrawal. Expression of Nestin, NeuN, MAP2, and GFAP in ADSCs and terminally differentiated neurospheres was shown by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting, and immunocytochemistry; cell proliferation in neurospheres was evaluated by cell cycle analyses, immunostaining, and flow cytometry. These data strongly support the conclusion that human ADSCs can successfully differentiate into neurospheres efficiently on uncoated culture flasks, which present similar molecular marker pattern and proliferative ability with NSCs derived from embryonic and adult brain tissues. Therefore, human ADSCs may be an ideal alternative source of stem cells for the treatment of neurodegenerative diseases.
Collapse
|
20
|
Cottenie E, Kochanski A, Jordanova A, Bansagi B, Zimon M, Horga A, Jaunmuktane Z, Saveri P, Rasic VM, Baets J, Bartsakoulia M, Ploski R, Teterycz P, Nikolic M, Quinlivan R, Laura M, Sweeney MG, Taroni F, Lunn MP, Moroni I, Gonzalez M, Hanna MG, Bettencourt C, Chabrol E, Franke A, von Au K, Schilhabel M, Kabzińska D, Hausmanowa-Petrusewicz I, Brandner S, Lim SC, Song H, Choi BO, Horvath R, Chung KW, Zuchner S, Pareyson D, Harms M, Reilly MM, Houlden H. Truncating and missense mutations in IGHMBP2 cause Charcot-Marie Tooth disease type 2. Am J Hum Genet 2014; 95:590-601. [PMID: 25439726 PMCID: PMC4225647 DOI: 10.1016/j.ajhg.2014.10.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/01/2014] [Indexed: 11/18/2022] Open
Abstract
Using a combination of exome sequencing and linkage analysis, we investigated an English family with two affected siblings in their 40s with recessive Charcot-Marie Tooth disease type 2 (CMT2). Compound heterozygous mutations in the immunoglobulin-helicase-μ-binding protein 2 (IGHMBP2) gene were identified. Further sequencing revealed a total of 11 CMT2 families with recessively inherited IGHMBP2 gene mutations. IGHMBP2 mutations usually lead to spinal muscular atrophy with respiratory distress type 1 (SMARD1), where most infants die before 1 year of age. The individuals with CMT2 described here, have slowly progressive weakness, wasting and sensory loss, with an axonal neuropathy typical of CMT2, but no significant respiratory compromise. Segregating IGHMBP2 mutations in CMT2 were mainly loss-of-function nonsense in the 5' region of the gene in combination with a truncating frameshift, missense, or homozygous frameshift mutations in the last exon. Mutations in CMT2 were predicted to be less aggressive as compared to those in SMARD1, and fibroblast and lymphoblast studies indicate that the IGHMBP2 protein levels are significantly higher in CMT2 than SMARD1, but lower than controls, suggesting that the clinical phenotype differences are related to the IGHMBP2 protein levels.
Collapse
Affiliation(s)
- Ellen Cottenie
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Molecular Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Andrzej Kochanski
- Neuromuscular Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Centre of Biostructure, Medical University of Warsaw, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Albena Jordanova
- VIB Department of Molecular Genetics, University of Antwerp, Antwerpen 2610, Belgium
| | - Boglarka Bansagi
- Institute of Genetic Medicine, MRC Centre for Neuromuscular Diseases, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Magdalena Zimon
- VIB Department of Molecular Genetics, University of Antwerp, Antwerpen 2610, Belgium
| | - Alejandro Horga
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Molecular Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Zane Jaunmuktane
- Division of Neuropathology and Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Paola Saveri
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, IRCCS Foundation, C. Besta Neurological Institute, Via Celoria 11, 20133 Milan, Italy
| | - Vedrana Milic Rasic
- Clinic for Neurology and Psychiatry for Children and Youth, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Jonathan Baets
- VIB Department of Molecular Genetics, University of Antwerp, Antwerpen 2610, Belgium; Laboratory of Neurogenetics, University of Antwerp, Antwerpen 2610, Belgium; Department of Neurology, Antwerp University Hospital, Antwerpen, Belgium
| | - Marina Bartsakoulia
- Institute of Genetic Medicine, MRC Centre for Neuromuscular Diseases, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Rafal Ploski
- Department of Medical Genetics, Centre of Biostructure, Medical University of Warsaw, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Pawel Teterycz
- Department of Medical Genetics, Centre of Biostructure, Medical University of Warsaw, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Milos Nikolic
- University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia
| | - Ros Quinlivan
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Matilde Laura
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Molecular Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Mary G Sweeney
- Neurogenetics Laboratory, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Franco Taroni
- Unit of Genetics of Neurodegenerative and Metabolic Disease IRCCS Foundation, C. Besta Neurological Institute, Via Celoria 11, 20133 Milan, Italy
| | - Michael P Lunn
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Isabella Moroni
- Child Neurology Unit, IRCCS Foundation, C. Besta Neurological Institute, Via Celoria 11, 20133 Milan, Italy
| | - Michael Gonzalez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, FL 33136, USA
| | - Michael G Hanna
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Molecular Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Conceicao Bettencourt
- Department of Molecular Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Elodie Chabrol
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Andre Franke
- Christian-Albrechts-University, 24118 Kiel, Germany
| | - Katja von Au
- SPZ Pediatric Neurology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | | | - Dagmara Kabzińska
- Neuromuscular Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Centre of Biostructure, Medical University of Warsaw, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Irena Hausmanowa-Petrusewicz
- Neuromuscular Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Centre of Biostructure, Medical University of Warsaw, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Sebastian Brandner
- Division of Neuropathology and Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Siew Choo Lim
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673
| | - Haiwei Song
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673; Life Sciences Institute, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Byung-Ok Choi
- Department of Neurology, Sungkyunkwan University School of Medicine, Seoul 137-710, Korea
| | - Rita Horvath
- Institute of Genetic Medicine, MRC Centre for Neuromuscular Diseases, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Ki-Wha Chung
- Department of Biological Science, Kongju National University, Chungnam 134-701, Korea
| | - Stephan Zuchner
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, FL 33136, USA
| | - Davide Pareyson
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, IRCCS Foundation, C. Besta Neurological Institute, Via Celoria 11, 20133 Milan, Italy
| | - Matthew Harms
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mary M Reilly
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Molecular Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Henry Houlden
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Molecular Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Neurogenetics Laboratory, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
21
|
Faravelli I, Frattini E, Ramirez A, Stuppia G, Nizzardo M, Corti S. iPSC-Based Models to Unravel Key Pathogenetic Processes Underlying Motor Neuron Disease Development. J Clin Med 2014; 3:1124-45. [PMID: 26237595 PMCID: PMC4470174 DOI: 10.3390/jcm3041124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 09/19/2014] [Accepted: 09/22/2014] [Indexed: 12/12/2022] Open
Abstract
Motor neuron diseases (MNDs) are neuromuscular disorders affecting rather exclusively upper motor neurons (UMNs) and/or lower motor neurons (LMNs). The clinical phenotype is characterized by muscular weakness and atrophy leading to paralysis and almost invariably death due to respiratory failure. Adult MNDs include sporadic and familial amyotrophic lateral sclerosis (sALS-fALS), while the most common infantile MND is represented by spinal muscular atrophy (SMA). No effective treatment is ccurrently available for MNDs, as for the vast majority of neurodegenerative disorders, and cures are limited to supportive care and symptom relief. The lack of a deep understanding of MND pathogenesis accounts for the difficulties in finding a cure, together with the scarcity of reliable in vitro models. Recent progresses in stem cell field, in particular in the generation of induced Pluripotent Stem Cells (iPSCs) has made possible for the first time obtaining substantial amounts of human cells to recapitulate in vitro some of the key pathogenetic processes underlying MNDs. In the present review, recently published studies involving the use of iPSCs to unravel aspects of ALS and SMA pathogenesis are discussed with an overview of their implications in the process of finding a cure for these still orphan disorders.
Collapse
Affiliation(s)
- Irene Faravelli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Emanuele Frattini
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Agnese Ramirez
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Giulia Stuppia
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Monica Nizzardo
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
22
|
LoMauro A, Romei M, Priori R, Laviola M, D’Angelo MG, Aliverti A. Alterations of thoraco-abdominal volumes and asynchronies in patients with spinal muscle atrophy type III. Respir Physiol Neurobiol 2014; 197:1-8. [DOI: 10.1016/j.resp.2014.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 11/29/2022]
|
23
|
Sumner C, d’Ydewalle C, Wooley J, Fawcett K, Hernandez D, Gardiner A, Kalmar B, Baloh R, Gonzalez M, Züchner S, Stanescu H, Kleta R, Mankodi A, Cornblath D, Boylan K, Reilly M, Greensmith L, Singleton A, Harms M, Rossor A, Houlden H. A dominant mutation in FBXO38 causes distal spinal muscular atrophy with calf predominance. Am J Hum Genet 2013; 93:976-83. [PMID: 24207122 PMCID: PMC3824115 DOI: 10.1016/j.ajhg.2013.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 11/19/2022] Open
Abstract
Spinal muscular atrophies (SMAs) are a heterogeneous group of inherited disorders characterized by degeneration of anterior horn cells and progressive muscle weakness. In two unrelated families affected by a distinct form of autosomal-dominant distal SMA initially manifesting with calf weakness, we identified by genetic linkage analysis and exome sequencing a heterozygous missense mutation, c.616T>C (p.Cys206Arg), in F-box protein 38 (FBXO38). FBXO38 is a known coactivator of the transcription factor Krüppel-like factor 7 (KLF7), which regulates genes required for neuronal axon outgrowth and repair. The p.Cys206Arg substitution did not alter the subcellular localization of FBXO38 but did impair KLF7-mediated transactivation of a KLF7-responsive promoter construct and endogenous KLF7 target genes in both heterologously expressing human embryonic kidney 293T cells and fibroblasts derived from individuals with the FBXO38 missense mutation. This transcriptional dysregulation was associated with an impairment of neurite outgrowth in primary motor neurons. Together, these results suggest that a transcriptional regulatory pathway that has a well-established role in axonal development could also be critical for neuronal maintenance and highlight the importance of FBXO38 and KLF7 activity in motor neurons.
Collapse
Affiliation(s)
- Charlotte J. Sumner
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Constantin d’Ydewalle
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Laboratory for Neurobiology, Vesalius Research Center, VIB and KU Leuven, 3000 Leuven, Belgium
| | - Joe Wooley
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Katherine A. Fawcett
- Department of Molecular Neuroscience, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The MRC Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alice R. Gardiner
- Department of Molecular Neuroscience, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The MRC Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Bernadett Kalmar
- The MRC Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Robert H. Baloh
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90095, USA
| | - Michael Gonzalez
- Dr. John T. MacDonald Department of Human Genetics and Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Stephan Züchner
- Dr. John T. MacDonald Department of Human Genetics and Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Horia C. Stanescu
- Center for Nephrology, University College London, London WC1N 3BG, UK
| | - Robert Kleta
- Center for Nephrology, University College London, London WC1N 3BG, UK
| | - Ami Mankodi
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - David R. Cornblath
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kevin B. Boylan
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Mary M. Reilly
- Department of Molecular Neuroscience, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The MRC Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Linda Greensmith
- The MRC Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Andrew B. Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew B. Harms
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander M. Rossor
- The MRC Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Henry Houlden
- Department of Molecular Neuroscience, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The MRC Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
24
|
Dysregulation of synaptogenesis genes antecedes motor neuron pathology in spinal muscular atrophy. Proc Natl Acad Sci U S A 2013; 110:19348-53. [PMID: 24191055 DOI: 10.1073/pnas.1319280110] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The motor neuron (MN) degenerative disease, spinal muscular atrophy (SMA) is caused by deficiency of SMN (survival motor neuron), a ubiquitous and indispensable protein essential for biogenesis of snRNPs, key components of pre-mRNA processing. However, SMA's hallmark MN pathology, including neuromuscular junction (NMJ) disruption and sensory-motor circuitry impairment, remains unexplained. Toward this end, we used deep RNA sequencing (RNA-seq) to determine if there are any transcriptome changes in MNs and surrounding spinal cord glial cells (white matter, WM) microdissected from SMN-deficient SMA mouse model at presymptomatic postnatal day 1 (P1), before detectable MN pathology (P4-P5). The RNA-seq results, previously unavailable for SMA at any stage, revealed cell-specific selective mRNA dysregulations (~300 of 11,000 expressed genes in each, MN and WM), many of which are known to impair neurons. Remarkably, these dysregulations include complete skipping of agrin's Z exons, critical for NMJ maintenance, strong up-regulation of synapse pruning-promoting complement factor C1q, and down-regulation of Etv1/ER81, a transcription factor required for establishing sensory-motor circuitry. We propose that dysregulation of such specific MN synaptogenesis genes, compounded by many additional transcriptome abnormalities in MNs and WM, link SMN deficiency to SMA's signature pathology.
Collapse
|
25
|
Hirano M, Kato S, Kobayashi K, Okada T, Yaginuma H, Kobayashi K. Highly efficient retrograde gene transfer into motor neurons by a lentiviral vector pseudotyped with fusion glycoprotein. PLoS One 2013; 8:e75896. [PMID: 24086660 PMCID: PMC3782444 DOI: 10.1371/journal.pone.0075896] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/18/2013] [Indexed: 12/14/2022] Open
Abstract
The development of gene therapy techniques to introduce transgenes that promote neuronal survival and protection provides effective therapeutic approaches for neurological and neurodegenerative diseases. Intramuscular injection of adenoviral and adeno-associated viral vectors, as well as lentiviral vectors pseudotyped with rabies virus glycoprotein (RV-G), permits gene delivery into motor neurons in animal models for motor neuron diseases. Recently, we developed a vector with highly efficient retrograde gene transfer (HiRet) by pseudotyping a human immunodeficiency virus type 1 (HIV-1)-based vector with fusion glycoprotein B type (FuG-B) or a variant of FuG-B (FuG-B2), in which the cytoplasmic domain of RV-G was replaced by the corresponding part of vesicular stomatitis virus glycoprotein (VSV-G). We have also developed another vector showing neuron-specific retrograde gene transfer (NeuRet) with fusion glycoprotein C type, in which the short C-terminal segment of the extracellular domain and transmembrane/cytoplasmic domains of RV-G was substituted with the corresponding regions of VSV-G. These two vectors afford the high efficiency of retrograde gene transfer into different neuronal populations in the brain. Here we investigated the efficiency of the HiRet (with FuG-B2) and NeuRet vectors for retrograde gene transfer into motor neurons in the spinal cord and hindbrain in mice after intramuscular injection and compared it with the efficiency of the RV-G pseudotype of the HIV-1-based vector. The main highlight of our results is that the HiRet vector shows the most efficient retrograde gene transfer into both spinal cord and hindbrain motor neurons, offering its promising use as a gene therapeutic approach for the treatment of motor neuron diseases.
Collapse
Affiliation(s)
- Miyabi Hirano
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute of Physiological Sciences, Okazaki, Japan
| | - Tomoaki Okada
- Department of Neuroanatomy & Embryology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroyuki Yaginuma
- Department of Neuroanatomy & Embryology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan
- * E-mail:
| |
Collapse
|
26
|
Neveling K, Martinez-Carrera L, Hölker I, Heister A, Verrips A, Hosseini-Barkooie S, Gilissen C, Vermeer S, Pennings M, Meijer R, te Riele M, Frijns C, Suchowersky O, MacLaren L, Rudnik-Schöneborn S, Sinke R, Zerres K, Lowry R, Lemmink H, Garbes L, Veltman J, Schelhaas H, Scheffer H, Wirth B. Mutations in BICD2, which encodes a golgin and important motor adaptor, cause congenital autosomal-dominant spinal muscular atrophy. Am J Hum Genet 2013; 92:946-54. [PMID: 23664116 DOI: 10.1016/j.ajhg.2013.04.011] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/15/2013] [Accepted: 04/15/2013] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a heterogeneous group of neuromuscular disorders caused by degeneration of lower motor neurons. Although functional loss of SMN1 is associated with autosomal-recessive childhood SMA, the genetic cause for most families affected by dominantly inherited SMA is unknown. Here, we identified pathogenic variants in bicaudal D homolog 2 (Drosophila) (BICD2) in three families afflicted with autosomal-dominant SMA. Affected individuals displayed congenital slowly progressive muscle weakness mainly of the lower limbs and congenital contractures. In a large Dutch family, linkage analysis identified a 9q22.3 locus in which exome sequencing uncovered c.320C>T (p.Ser107Leu) in BICD2. Sequencing of 23 additional families affected by dominant SMA led to the identification of pathogenic variants in one family from Canada (c.2108C>T [p.Thr703Met]) and one from the Netherlands (c.563A>C [p.Asn188Thr]). BICD2 is a golgin and motor-adaptor protein involved in Golgi dynamics and vesicular and mRNA transport. Transient transfection of HeLa cells with all three mutant BICD2 cDNAs caused massive Golgi fragmentation. This observation was even more prominent in primary fibroblasts from an individual harboring c.2108C>T (p.Thr703Met) (affecting the C-terminal coiled-coil domain) and slightly less evident in individuals with c.563A>C (p.Asn188Thr) (affecting the N-terminal coiled-coil domain). Furthermore, BICD2 levels were reduced in affected individuals and trapped within the fragmented Golgi. Previous studies have shown that Drosophila mutant BicD causes reduced larvae locomotion by impaired clathrin-mediated synaptic endocytosis in neuromuscular junctions. These data emphasize the relevance of BICD2 in synaptic-vesicle recycling and support the conclusion that BICD2 mutations cause congenital slowly progressive dominant SMA.
Collapse
|
27
|
Prather RS, Lorson M, Ross JW, Whyte JJ, Walters E. Genetically engineered pig models for human diseases. Annu Rev Anim Biosci 2013; 1:203-19. [PMID: 25387017 DOI: 10.1146/annurev-animal-031412-103715] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although pigs are used widely as models of human disease, their utility as models has been enhanced by genetic engineering. Initially, transgenes were added randomly to the genome, but with the application of homologous recombination, zinc finger nucleases, and transcription activator-like effector nuclease (TALEN) technologies, now most any genetic change that can be envisioned can be completed. To date these genetic modifications have resulted in animals that have the potential to provide new insights into human diseases for which a good animal model did not exist previously. These new animal models should provide the preclinical data for treatments that are developed for diseases such as Alzheimer's disease, cystic fibrosis, retinitis pigmentosa, spinal muscular atrophy, diabetes, and organ failure. These new models will help to uncover aspects and treatments of these diseases that were otherwise unattainable. The focus of this review is to describe genetically engineered pigs that have resulted in models of human diseases.
Collapse
Affiliation(s)
- Randall S Prather
- Division of Animal Science, National Swine Resource and Research Center, and
| | | | | | | | | |
Collapse
|
28
|
Lee HLR, Dougherty JP. Pharmaceutical therapies to recode nonsense mutations in inherited diseases. Pharmacol Ther 2012; 136:227-66. [PMID: 22820013 DOI: 10.1016/j.pharmthera.2012.07.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2012] [Indexed: 12/21/2022]
Abstract
Nonsense codons, generated from nonsense mutations or frameshifts, contribute significantly to the spectrum of inherited human diseases such as cystic fibrosis, Duchenne muscular dystrophy, hemophilia, spinal muscular atrophy, and many forms of cancer. The presence of a mutant nonsense codon results in premature termination to preclude the synthesis of a full-length protein and leads to aberrations in gene expression. Suppression therapy to recode a premature termination codon with an amino acid allowing readthrough to rescue the production of a full-length protein presents a promising strategy for treatment of patients suffering from debilitating nonsense-mediated disorders. Suppression therapy using aminoglycosides to promote readthrough in vitro have been known since the sixties. Recent progress in the field of recoding via pharmaceuticals has led to the continuous discovery and development of several pharmacological agents with nonsense suppression activities. Here, we review the mechanisms that are involved in discriminating normal versus premature termination codons, the factors involved in readthrough efficiency, the epidemiology of several well-known nonsense-mediated diseases, and the various pharmacological agents (aminoglycoside and non-aminoglycoside compounds) that are currently being employed in nonsense suppression therapy studies. We also discuss how these therapeutic agents can be used to regulate gene expression for gene therapy applications.
Collapse
Affiliation(s)
- Hui-Ling Rose Lee
- Department of Molecular Genetics, Microbiology, and Immunology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ, USA
| | | |
Collapse
|
29
|
Satoh M, Ceribelli A, Chan EKL. Common pathways of autoimmune inflammatory myopathies and genetic neuromuscular disorders. Clin Rev Allergy Immunol 2012; 42:16-25. [PMID: 22083460 DOI: 10.1007/s12016-011-8286-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
It has been shown that many hereditary motor neuron diseases are caused by mutation of RNA processing enzymes. Survival of motor neuron 1 (SMN1) is well-known as a causative gene for spinal muscular atrophy (SMA) and mutations of glycyl- and tyrosyl-tRNA synthetases are identified as a cause of distal SMA and Charcot-Marie-Tooth disease. Why and how the dysfunction of these ubiquitously expressed genes involved in RNA processing can cause a specific neurological disorder is not well understood. Interestingly, SMN complex has been identified recently as a new target of autoantibodies in polymyositis (PM). Autoantibodies in systemic rheumatic diseases are clinically useful biomarkers associated with a particular diagnosis, subset of a disease, or certain clinical characteristics. Many autoantibodies produced in patients with polymyositis/dermatomyositis (PM/DM) target RNA-protein complexes such as aminoacyl tRNA synthetases. It is interesting to note these same RNA-protein complexes recognized by autoantibodies in PM/DM are also responsible for genetic neuromuscular disease. Certain RNA-protein complexes are also targets of autoantibodies in paraneoplastic neurological disorders. Thus, there are several interesting associations between RNA-processing enzymes and neuromuscular disorders. Although pathogenetic roles of autoantibodies to intracellular antigens are generally considered unlikely, understanding the mechanisms of antigen selection in a particular disease and specific neurological symptoms caused by disruption of ubiquitous RNA-processing enzyme may help identify a common path in genetic neuromuscular disorders and autoimmunity in inflammatory myopathies.
Collapse
Affiliation(s)
- Minoru Satoh
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Florida, Gainesville, FL 32610-0221, USA.
| | | | | |
Collapse
|
30
|
Functional neuromuscular junctions formed by embryonic stem cell-derived motor neurons. PLoS One 2012; 7:e36049. [PMID: 22574134 PMCID: PMC3344836 DOI: 10.1371/journal.pone.0036049] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 03/26/2012] [Indexed: 01/21/2023] Open
Abstract
A key objective of stem cell biology is to create physiologically relevant cells suitable for modeling disease pathologies in vitro. Much progress towards this goal has been made in the area of motor neuron (MN) disease through the development of methods to direct spinal MN formation from both embryonic and induced pluripotent stem cells. Previous studies have characterized these neurons with respect to their molecular and intrinsic functional properties. However, the synaptic activity of stem cell-derived MNs remains less well defined. In this study, we report the development of low-density co-culture conditions that encourage the formation of active neuromuscular synapses between stem cell-derived MNs and muscle cells in vitro. Fluorescence microscopy reveals the expression of numerous synaptic proteins at these contacts, while dual patch clamp recording detects both spontaneous and multi-quantal evoked synaptic responses similar to those observed in vivo. Together, these findings demonstrate that stem cell-derived MNs innervate muscle cells in a functionally relevant manner. This dual recording approach further offers a sensitive and quantitative assay platform to probe disorders of synaptic dysfunction associated with MN disease.
Collapse
|
31
|
Abstract
Bench to bedside progress has been widely anticipated for a growing number of neurodegenerative disorders. Of these, spinal muscular atrophy (SMA) is perhaps the best poised to capitalize on advances in targeted therapeutics development over the next few years. Several laboratories have achieved compelling success in SMA animal models using sophisticated methods for targeted delivery, repair, or increased expression of the survival motor neuron protein, SMN. The clinical community is actively collaborating to identify, develop, and validate outcome measures and biomarkers in parallel with laboratory efforts. Innovative trial design and synergistic approaches to maximize proactive care in conjunction with treatment with one or more of the promising pharmacologic and biologic therapies currently in the pipeline will maximize our chances to achieve meaningful outcomes for patients. This review highlights recent promising scientific and clinical advances bringing us ever closer to effective treatment(s) for our patients with SMA.
Collapse
|
32
|
Fiorillo C, Moro F, Brisca G, Astrea G, Nesti C, Bálint Z, Olschewski A, Meschini MC, Guelly C, Auer-Grumbach M, Battini R, Pedemonte M, Romano A, Menchise V, Biancheri R, Santorelli FM, Bruno C. TRPV4 mutations in children with congenital distal spinal muscular atrophy. Neurogenetics 2012; 13:195-203. [PMID: 22526352 DOI: 10.1007/s10048-012-0328-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 03/29/2012] [Indexed: 01/15/2023]
Abstract
Inherited disorders characterized by motor neuron loss and muscle weakness are genetically heterogeneous. The recent identification of mutations in the gene encoding transient receptor potential vanilloid 4 (TRPV4) in distal spinal muscular atrophy (dSMA) prompted us to screen for TRPV4 mutations in a small group of children with compatible phenotype. In a girl with dSMA and vocal cord paralysis, we detected a new variant (p.P97R) localized in the cytosolic N-terminus of the TRPV4 protein, upstream of the ankyrin-repeat domain, where the great majority of disease-associated mutations reside. In another child with congenital dSMA, in this case associated with bone abnormalities, we detected a previously reported mutation (p.R232C). Functional analysis of the novel p.P97R mutation in a heterologous system demonstrated a loss-of-function mechanism. Protein localization studies in muscle, skin, and cultured skin fibroblasts from both patients showed normal protein expression. No TRPV4 mutations were detected in four children with dSMA without bone or vocal cord involvement. Adding to the clinical and molecular heterogeneity of TRPV4-associated diseases, our results suggest that molecular testing of the TRPV4 gene is warranted in cases of congenital dSMA with bone abnormalities and vocal cord paralysis.
Collapse
Affiliation(s)
- Chiara Fiorillo
- Neuromuscular Unit, IRCCS Stella Maris, Via dei Giacinti 2, 56028 Pisa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lorusso ML, Civati F, Molteni M, Turconi AC, Bresolin N, D'Angelo MG. Specific profiles of neurocognitive and reading functions in a sample of 42 Italian boys with Duchenne Muscular Dystrophy. Child Neuropsychol 2012; 19:350-69. [PMID: 22385039 DOI: 10.1080/09297049.2012.660912] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
A group of 42 Italian boys with Duchenne Muscular Dystrophy was compared with a control group of 10 boys with Spinal Muscular Atrophy and Osteogenesis Imperfecta on tests assessing general intellectual ability, language, neuropsychological functions, and reading skills with the aim of describing a comprehensive profile of the various functions and investigating their interrelationships. The influence of general intellectual level on performance was analyzed. Further, correlations between various neuropsychological measures and language performances were computed for the group with Duchenne Muscular Dystrophy, as well as the correlations between reading scores and other cognitive and linguistic measures. A general lowering in VIQ, PIQ, and FSIQ scores was found to characterize the group with Duchenne Muscular Dystrophy. Expressive language skills were within the normal range, while syntactic and grammatical comprehension were significantly impaired. The presence of below-average reading performances was further confirmed. However, unlike previous studies on irregular orthographies, the present results show that (a) the mild reading difficulties found in the sample essentially concern speed rather than accuracy; (b) they concern word rather than nonword reading; (c) lower reading performances are related to lower scores in general IQ; (d) no correlations emerge with phonological abilities, verbal short-term memory, or working memory, but rather with long-term memory and lexical skills. This may suggest that language-specific effects modulate the cognitive expressions of Duchenne Muscular Dystrophy and raises the possibility that the dysfunctions underlying the reading difficulties observed in affected readers of regular orthographies involve different neurocognitive systems than the cortico-cerebellar circuits usually invoked.
Collapse
Affiliation(s)
- Maria Luisa Lorusso
- Department of Neuro-Rehabilitation-II, Unit of Neuropsychology of Developmental Disorders, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Lecco, Italy.
| | | | | | | | | | | |
Collapse
|
34
|
Ekici B, Bozkurt B, Tatlı B, Calışkan M, Aydınlı N, Ozmen M. Demographic characteristics of SMA type 1 patients at a tertiary center in Turkey. Eur J Pediatr 2012; 171:549-52. [PMID: 22016262 DOI: 10.1007/s00431-011-1607-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 10/07/2011] [Indexed: 12/01/2022]
Abstract
UNLABELLED The aim of this study was to demonstrate demographics of 39 consecutive Spinal Muscular Atrophy (SMA) type 1 patients diagnosed genetically in a tertiary center between June 2006 and June 2009. There was history of consanguineous marriage in 27 (69%) patients. The average patient lifespan was 251 days (30-726 days). The average patient age at diagnosis was 129 days (33-297 days). A statistically significant correlation was found between the age at diagnosis and the lifespan (p = 0.00). No significant correlation was found between the time spent in intensive care and the lifespan (p = 0.43). Routine physical therapy was found to have no significant impact on the lifespan average (p = 0.17). The cause of death in all of our patients was respiratory issues. Genetic counseling was given to 35 families. A second child with SMA was born in three out of the 14 families who declined prenatal diagnosis. CONCLUSION A national program is needed in Turkey for SMA prevention and creation of expert teams for the management of these patients.
Collapse
Affiliation(s)
- Barış Ekici
- Department of Pediatric Neurology, Istanbul Medical Faculty, Ortaköy Dereboyu cad. Arkeon sitesi A 5 blok D 3, Beşiktaş, Fatih, Istanbul, Turkey.
| | | | | | | | | | | |
Collapse
|
35
|
Neurocognitive profiles in Duchenne muscular dystrophy and gene mutation site. Pediatr Neurol 2011; 45:292-9. [PMID: 22000308 PMCID: PMC3200430 DOI: 10.1016/j.pediatrneurol.2011.08.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 08/08/2011] [Indexed: 11/23/2022]
Abstract
The presence of nonprogressive cognitive impairment is recognized as a common feature in a substantial proportion of patients with Duchenne muscular dystrophy. To investigate the possible role of mutations along the dystrophin gene affecting different brain dystrophin isoforms and specific cognitive profiles, 42 school-age children affected with Duchenne muscular dystrophy, subdivided according to sites of mutations along the dystrophin gene, underwent a battery of tests tapping a wide range of intellectual, linguistic, and neuropsychologic functions. Full-scale intelligence quotient was approximately 1 S.D. below the population average in the whole group of dystrophic children. Patients with Duchenne muscular dystrophy and mutations located in the distal portion of the dystrophin gene (involving the 140-kDa brain protein isoform, called Dp140) were generally more severely affected and expressed different patterns of strengths and impairments, compared with patients with Duchenne muscular dystrophy and mutations located in the proximal portion of the dystrophin gene (not involving Dp140). Patients with Duchenne muscular dystrophy and distal mutations demonstrated specific impairments in visuospatial functions and visual memory (which seemed intact in proximally mutated patients) and greater impairment in syntactic processing.
Collapse
|
36
|
Abstract
Over the past 20 years, stem cell technologies have become an increasingly attractive option to investigate and treat neurodegenerative diseases. In the current review, we discuss the process of extending basic stem cell research into translational therapies for patients suffering from neurodegenerative diseases. We begin with a discussion of the burden of these diseases on society, emphasizing the need for increased attention toward advancing stem cell therapies. We then explain the various types of stem cells utilized in neurodegenerative disease research, and outline important issues to consider in the transition of stem cell therapy from bench to bedside. Finally, we detail the current progress regarding the applications of stem cell therapies to specific neurodegenerative diseases, focusing on Parkinson disease, Huntington disease, Alzheimer disease, amyotrophic lateral sclerosis, and spinal muscular atrophy. With a greater understanding of the capacity of stem cell technologies, there is growing public hope that stem cell therapies will continue to progress into realistic and efficacious treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- J Simon Lunn
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | | | | | | |
Collapse
|
37
|
Ling KKY, Gibbs RM, Feng Z, Ko CP. Severe neuromuscular denervation of clinically relevant muscles in a mouse model of spinal muscular atrophy. Hum Mol Genet 2011; 21:185-95. [PMID: 21968514 DOI: 10.1093/hmg/ddr453] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Spinal muscular atrophy (SMA), a motoneuron disease caused by a deficiency of the survival of motor neuron (SMN) protein, is characterized by motoneuron loss and muscle weakness. It remains unclear whether widespread loss of neuromuscular junctions (NMJs) is involved in SMA pathogenesis. We undertook a systematic examination of NMJ innervation patterns in >20 muscles in the SMNΔ7 SMA mouse model. We found that severe denervation (<50% fully innervated endplates) occurs selectively in many vulnerable axial muscles and several appendicular muscles at the disease end stage. Since these vulnerable muscles were located throughout the body and were comprised of varying muscle fiber types, it is unlikely that muscle location or fiber type determines susceptibility to denervation. Furthermore, we found a similar extent of neurofilament accumulation at NMJs in both vulnerable and resistant muscles before the onset of denervation, suggesting that neurofilament accumulation does not predict subsequent NMJ denervation. Since vulnerable muscles were initially innervated, but later denervated, loss of innervation in SMA may be attributed to defects in synapse maintenance. Finally, we found that denervation was amendable by trichostatin A (TSA) treatment, which increased innervation in clinically relevant muscles in TSA-treated SMNΔ7 mice. Our findings suggest that neuromuscular denervation in vulnerable muscles is a widespread pathology in SMA, and can serve as a preparation for elucidating the biological basis of synapse loss, and for evaluating therapeutic efficacy.
Collapse
Affiliation(s)
- Karen K Y Ling
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2520, USA
| | | | | | | |
Collapse
|
38
|
Sanz-García M, Vázquez-Cedeira M, Kellerman E, Renbaum P, Levy-Lahad E, Lazo PA. Substrate profiling of human vaccinia-related kinases identifies coilin, a Cajal body nuclear protein, as a phosphorylation target with neurological implications. J Proteomics 2011; 75:548-60. [PMID: 21920476 DOI: 10.1016/j.jprot.2011.08.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/19/2011] [Accepted: 08/23/2011] [Indexed: 01/13/2023]
Abstract
Protein phosphorylation by kinases plays a central role in the regulation and coordination of multiple biological processes. In general, knowledge on kinase specificity is restricted to substrates identified in the context of specific cellular responses, but kinases are likely to have multiple additional substrates and be integrated in signaling networks that might be spatially and temporally different, and in which protein complexes and subcellular localization can play an important role. In this report the substrate specificity of atypical human vaccinia-related kinases (VRK1 and VRK2) using a human peptide-array containing 1080 sequences phosphorylated in known signaling pathways has been studied. The two kinases identify a subset of potential peptide targets, all of them result in a consensus sequence composed of at least four basic residues in peptide targets. Linear peptide arrays are therefore a useful approach in the characterization of kinases and substrate identification, which can contribute to delineate the signaling network in which VRK proteins participate. One of these target proteins is coilin; a basic protein located in nuclear Cajal bodies. Coilin is phosphorylated in Ser184 by both VRK1 and VRK2. Coilin colocalizes and interacts with VRK1 in Cajal bodies, but not with the mutant VRK1 (R358X). VRK1 (R358X) is less active than VRK1. Altered regulation of coilin might be implicated in several neurological diseases such as ataxias and spinal muscular atrophies.
Collapse
Affiliation(s)
- Marta Sanz-García
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas(CSIC)-Universidad de Salamanca, Salamanca 37007, Spain
| | | | | | | | | | | |
Collapse
|
39
|
Lunn JS, Sakowski SA, Federici T, Glass JD, Boulis NM, Feldman EL. Stem cell technology for the study and treatment of motor neuron diseases. Regen Med 2011; 6:201-13. [PMID: 21391854 DOI: 10.2217/rme.11.6] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis and spinal muscular atrophy are devastating neurodegenerative diseases that lead to the specific loss of motor neurons. Recently, stem cell technologies have been developed for the investigation and treatment of both diseases. Here we discuss the different stem cells currently being studied for mechanistic discovery and therapeutic development, including embryonic, adult and induced pluripotent stem cells. We also present supporting evidence for the utilization of stem cell technology in the treatment of amyotrophic lateral sclerosis and spinal muscular atrophy, and describe key issues that must be considered for the transition of stem cell therapies for motor neuron diseases from bench to bedside. Finally, we discuss the first-in-human Phase I trial currently underway examining the safety and feasibility of intraspinal stem cell injections in amyotrophic lateral sclerosis patients as a foundation for translating stem cell therapies for various neurological diseases.
Collapse
Affiliation(s)
- J Simon Lunn
- University of Michigan Department of Neurology, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
40
|
Singh NN, Singh RN. Alternative splicing in spinal muscular atrophy underscores the role of an intron definition model. RNA Biol 2011; 8:600-6. [PMID: 21654213 DOI: 10.4161/rna.8.4.16224] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Humans have two nearly identical copies of the Survival Motor Neuron (SMN) gene: SMN1 and SMN2. The two SMN genes code for identical proteins; however, SMN2 predominantly generates a shorter transcript due to skipping of exon 7, the last coding exon. Skipping of SMN2 exon 7 leads to production of a truncated SMN protein that is highly unstable. The inability of SMN2 to compensate for the loss of SMN1 results in spinal muscular atrophy (SMA), the second most prevalent genetic cause of infant mortality. Since SMN2 is almost universally present in SMA patients, correction of SMN2 exon 7 splicing holds the promise for cure. Consistently, SMN2 exon 7 splicing has emerged as one of the best studied splicing systems in humans. The vast amount of recent literature provides a clue that SMN2 exon 7 splicing is regulated by an intron definition mechanism, which does not require cross-exon communication as prerequisite for exon inclusion. Our conclusion is based on the prominent role of intronic cis-elements, some of them have emerged as the frontrunners among potential therapeutic targets of SMA. Further, the widely expressed T-cell-restricted intracellular antigen-1 (TIA1), a member of the Q-rich domain containing RNA-binding proteins, has recently been found to regulate SMN exon 7 splicing by binding to intron 7 sequences away from the 5′ ss. These findings make a strong argument for an "intron definition model", according to which regulatory sequences within a downstream intron are capable of enforcing exon inclusion even in the absence of a defined upstream 3′ ss of an alternatively spliced exon.
Collapse
Affiliation(s)
- Natalia N Singh
- Department of Biomedical Sciences Iowa State University, Ames, IA, USA
| | | |
Collapse
|