1
|
Traynor BJ. The interneuron hypothesis of amyotrophic lateral sclerosis. Brain 2025; 148:1045-1046. [PMID: 40179249 PMCID: PMC11967792 DOI: 10.1093/brain/awaf104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Affiliation(s)
- Bryan J Traynor
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
van den Bos MAJ, Menon P, Pavey N, Higashihara M, Kiernan MC, Vucic S. Direct interrogation of cortical interneuron circuits in amyotrophic lateral sclerosis. Brain 2025; 148:1169-1179. [PMID: 39385724 DOI: 10.1093/brain/awae317] [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: 04/29/2024] [Revised: 07/30/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Cortical hyperexcitability is a key pathogenic feature of amyotrophic lateral sclerosis (ALS), believed to be mediated through complex interplay of cortical interneurons. To date, there has been no technological approach to facilitate the direct capture of cortical interneuron function. Through combination of transcranial magnetic stimulation (TMS) with advanced EEG, the present study examined GABAergic dysfunction in ALS by recording focused cortical output whilst applying TMS over the primary motor cortex contralateral to the site of symptom onset. Using both a single-pulse and a novel inhibitory paired-pulse paradigm, TMS-EEG studies were undertaken on 21 ALS patients and results compared with healthy controls. TMS responses captured by EEG form a discrete waveform known as the transcranial evoked potential (TEP), with positive (P) or upward deflections occurring at 30 (P30), 60 (P60) and 190 ms (P190) after TMS stimulus. Negative (N) or downward deflections occur at 44 (N44), 100 (N100) and 280 ms (N280) after TMS stimulus. The single-pulse TEPs recorded in ALS patients demonstrated novel differences suggestive of cortical GABAergic dysfunction. When compared with controls, the N100 component was significantly reduced (P < 0.05), whereas the P190 component increased (P < 0.05) in ALS patients. Additionally, the N44 component was correlated with muscle weakness (r = -0.501, P < 0.05). These findings were supported by reduced paired-pulse inhibition of TEP components in ALS patients (P60, P < 0.01; N100, P < 0.005), consistent with dysfunction of cortical interneuronal GABAA-ergic circuits. Furthermore, the reduction in short-interval intracortical inhibition, as reflected by changes in paired-pulse inhibition of the N100 component, was associated with longer disease duration in ALS patients (r = -0.698, P < 0.001). In conclusion, intensive and focused interrogation of the motor cortex using novel TMS-EEG combined technologies has established localized dysfunction of GABAergic circuits, supporting the notion that cortical hyperexcitability is mediated by cortical disinhibition in ALS. Dysfunction of GABAergic circuits was correlated with greater clinical disability and disease duration, implying pathophysiological significance.
Collapse
Affiliation(s)
- Mehdi A J van den Bos
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Parvathi Menon
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Nathan Pavey
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Matthew C Kiernan
- Neuroscience Research Australia, Randwick, Sydney, NSW 2031, Australia
- Institute of Neurological Sciences, Eastern Sydney Local Health District, Randwick, Sydney, NSW 2031, Australia
| | - Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| |
Collapse
|
3
|
Alzahrani AK, Imran M, Alshrari AS. Investigating the impact of SOD1 mutations on amyotrophic lateral sclerosis progression and potential drug repurposing through in silico analysis. J Biomol Struct Dyn 2024:1-16. [PMID: 39673548 DOI: 10.1080/07391102.2024.2439577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/29/2024] [Indexed: 12/16/2024]
Abstract
Superoxide dismutase 1 (SOD1) is a vital enzyme responsible for attenuating oxidative stress through its ability to facilitate the dismutation of the superoxide radical into oxygen and hydrogen peroxide. The progressive loss of motor neurons characterize amyotrophic lateral sclerosis (ALS), a crippling neurodegenerative disease that is caused by mutations in the SOD1 gene. In this study, in silico mutational analysis was performed to study the various mutations, the pathogenicity and stability ΔΔG (binding free energy) of the variant of SOD1. x in the protein variant analysis showed a considerable destabilizing effect with a ΔΔG value of -4.2 kcal/mol, signifying a notable impact on protein stability. Molecular dynamics simulations were conducted on both wild-type and C146R mutant SOD1. RMSD profiles indicated that both maintained consistent structural conformation over time. Additionally, virtual screening of 3067 FDA-approved drugs against the mutant SOD1 identified two potential binders, Tucatinib (51039094) and Regorafenib (11167602), which interacted with Leu106, similar to the control drug, Ebselen. Further simulations assessed the dynamic properties of SOD1 in monomeric and dimeric forms while bound to these compounds. 11167602 maintained stable interaction with the monomeric SOD1 mutant, whereas 51039094 and Ebselen dissociated from the monomeric protein's binding site. However, all three compounds were stably bound to the dimeric SOD1. MM/GBSA analysis revealed similar negative binding free energies for 11167602 and 51039094, identifying them as strong binders due to their interaction with Cys111. Experimental validation, including in vitro, cell-based, and in vivo assays are essential to confirm these candidates before advancing to clinical trials.
Collapse
Affiliation(s)
- A Khuzaim Alzahrani
- Department of Medical Laboratory Technology, Faculty of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Ahmed S Alshrari
- Department of Medical Laboratory Technology, Faculty of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
4
|
Saez-Atienzar S, Souza CDS, Chia R, Beal SN, Lorenzini I, Huang R, Levy J, Burciu C, Ding J, Gibbs JR, Jones A, Dewan R, Pensato V, Peverelli S, Corrado L, van Vugt JJFA, van Rheenen W, Tunca C, Bayraktar E, Xia M, Iacoangeli A, Shatunov A, Tiloca C, Ticozzi N, Verde F, Mazzini L, Kenna K, Al Khleifat A, Opie-Martin S, Raggi F, Filosto M, Piccinelli SC, Padovani A, Gagliardi S, Inghilleri M, Ferlini A, Vasta R, Calvo A, Moglia C, Canosa A, Manera U, Grassano M, Mandrioli J, Mora G, Lunetta C, Tanel R, Trojsi F, Cardinali P, Gallone S, Brunetti M, Galimberti D, Serpente M, Fenoglio C, Scarpini E, Comi GP, Corti S, Del Bo R, Ceroni M, Pinter GL, Taroni F, Bella ED, Bersano E, Curtis CJ, Lee SH, Chung R, Patel H, Morrison KE, Cooper-Knock J, Shaw PJ, Breen G, Dobson RJB, Dalgard CL, Scholz SW, Al-Chalabi A, van den Berg LH, McLaughlin R, Hardiman O, Cereda C, Sorarù G, D'Alfonso S, Chandran S, Pal S, Ratti A, Gellera C, Johnson K, Doucet-O'Hare T, Pasternack N, Wang T, Nath A, Siciliano G, Silani V, Başak AN, Veldink JH, Camu W, Glass JD, Landers JE, Chiò A, Sattler R, Shaw CE, Ferraiuolo L, Fogh I, Traynor BJ. Mechanism-free repurposing of drugs for C9orf72-related ALS/FTD using large-scale genomic data. CELL GENOMICS 2024; 4:100679. [PMID: 39437787 PMCID: PMC11605688 DOI: 10.1016/j.xgen.2024.100679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 09/22/2024] [Indexed: 10/25/2024]
Abstract
Repeat expansions in the C9orf72 gene are the most common genetic cause of (ALS) and frontotemporal dementia (FTD). Like other genetic forms of neurodegeneration, pinpointing the precise mechanism(s) by which this mutation leads to neuronal death remains elusive, and this lack of knowledge hampers the development of therapy for C9orf72-related disease. We used an agnostic approach based on genomic data (n = 41,273 ALS and healthy samples, and n = 1,516 C9orf72 carriers) to overcome these bottlenecks. Our drug-repurposing screen, based on gene- and expression-pattern matching and information about the genetic variants influencing onset age among C9orf72 carriers, identified acamprosate, a γ-aminobutyric acid analog, as a potentially repurposable treatment for patients carrying C9orf72 repeat expansions. We validated its neuroprotective effect in cell models and showed comparable efficacy to riluzole, the current standard of care. Our work highlights the potential value of genomics in repurposing drugs in situations where the underlying pathomechanisms are inherently complex. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Sara Saez-Atienzar
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Ohio State University, Columbus, OH 43210, USA.
| | - Cleide Dos Santos Souza
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ruth Chia
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Selina N Beal
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ileana Lorenzini
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Jennifer Levy
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Camelia Burciu
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - J Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Ashley Jones
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ramita Dewan
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Viviana Pensato
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Peverelli
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Lucia Corrado
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Joke J F A van Vugt
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ceren Tunca
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Elif Bayraktar
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Cinzia Tiloca
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Letizia Mazzini
- Amyotrophic Lateral Sclerosis Center, Department of Neurology "Maggiore della Carità" University Hospital, Novara, Italy
| | - Kevin Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Sarah Opie-Martin
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Flavia Raggi
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Massimiliano Filosto
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Cotti Piccinelli
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stella Gagliardi
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Maurizio Inghilleri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, 00185 Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Science, University of Ferrara, Ferrara, Italy
| | - Rosario Vasta
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Andrea Calvo
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Cristina Moglia
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Antonio Canosa
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Umberto Manera
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Maurizio Grassano
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Gabriele Mora
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Christian Lunetta
- Department of Neurorehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Institute of Milan, Milan, Italy; NEMO Clinical Center Milano, Fondazione Serena Onlus, Milan, Italy
| | - Raffaella Tanel
- Operative Unit of Neurology, S. Chiara Hospital, Trento, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | | | - Salvatore Gallone
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Maura Brunetti
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Maria Serpente
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Elio Scarpini
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo P Comi
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberto Del Bo
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Ceroni
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Giuseppe Lauria Pinter
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eleonora Dalla Bella
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
| | - Enrica Bersano
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; "L. Sacco" Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Charles J Curtis
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Sang Hyuck Lee
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Raymond Chung
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Hamel Patel
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Karen E Morrison
- School of Medicine, Dentistry, and Biomedical Sciences, Faculty of Medicine Health and Life Sciences, Queen's University, Belfast, UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Gerome Breen
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Richard J B Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK; NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Health Data Research UK London, University College London, London, UK; Institute of Health Informatics, University College London, London, UK; NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, London, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, King's College Hospital, London SE5 9RS, UK
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Russell McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Siddharthan Chandran
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Suvankar Pal
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; Centre for Neuroregeneration and Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Kory Johnson
- Bioinformatics Section, Information Technology Program (ITP), Division of Intramural Research (DIR), National Institute of Neurological Disorders & Stroke, NIH, Bethesda, MD 20892, USA
| | - Tara Doucet-O'Hare
- Neuro-oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Nicholas Pasternack
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Tongguang Wang
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Avindra Nath
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Ayşe Nazlı Başak
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - William Camu
- ALS Center, CHU Gui de Chauliac, University of Montpellier, Montpellier, France
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Rita Sattler
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christopher E Shaw
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Isabella Fogh
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK; National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; RNA Therapeutics Laboratory, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA.
| |
Collapse
|
5
|
Raffaele S, Nguyen N, Milanese M, Mannella FC, Boccazzi M, Frumento G, Bonanno G, Abbracchio MP, Bonifacino T, Fumagalli M. Montelukast improves disease outcome in SOD1 G93A female mice by counteracting oligodendrocyte dysfunction and aberrant glial reactivity. Br J Pharmacol 2024; 181:3303-3326. [PMID: 38751168 DOI: 10.1111/bph.16408] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/19/2024] [Accepted: 03/08/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by progressive motor neuron (MN) loss and consequent muscle atrophy, for which no effective therapies are available. Recent findings reveal that disease progression is fuelled by early aberrant neuroinflammation and the loss of oligodendrocytes with neuroprotective and remyelinating properties. On this basis, pharmacological interventions capable of restoring a pro-regenerative local milieu and re-establish proper oligodendrocyte functions may be beneficial. EXPERIMENTAL APPROACH Here, we evaluated the in vivo therapeutic effects of montelukast (MTK), an antagonist of the oligodendroglial G protein-coupled receptor 17 (GPR17) and of cysteinyl-leukotriene receptor 1 (CysLT1R) receptors on microglia and astrocytes, in the SOD1G93A ALS mouse model. We chronically treated SOD1G93A mice with MTK, starting from the early symptomatic disease stage. Disease progression was assessed by behavioural and immunohistochemical approaches. KEY RESULTS Oral MTK treatment significantly extended survival probability, delayed body weight loss and ameliorated motor functionalityonly in female SOD1G93A mice. Noteworthy, MTK significantly restored oligodendrocyte maturation and induced significant changes in the reactive phenotype and morphological features of microglia/macrophages and astrocytes in the spinal cord of female SOD1G93A mice, suggesting enhanced pro-regenerative functions. Importantly, concomitant MN preservation has been detected after MTK administration. No beneficial effects were observed in male mice, highlighting a sex-based difference in the protective activity of MTK. CONCLUSIONS AND IMPLICATIONS Our results provide the first preclinical evidence indicating that repurposing of MTK, a safe and marketed anti-asthmatic drug, may be a promising sex-specific strategy for personalized ALS treatment.
Collapse
Affiliation(s)
- Stefano Raffaele
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Nhung Nguyen
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca C Mannella
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Giulia Frumento
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
- Inter-University Center for the Promotion of the 3R Principles in Teaching and Research (Centro 3R), Pisa, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
6
|
Dash BP, Freischmidt A, Weishaupt JH, Hermann A. An integrative miRNA-mRNA expression analysis identifies miRNA signatures associated with SOD1 and TARDBP patient-derived motor neurons. Hum Mol Genet 2024; 33:1300-1314. [PMID: 38676626 DOI: 10.1093/hmg/ddae072] [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: 01/28/2024] [Revised: 03/27/2024] [Indexed: 04/29/2024] Open
Abstract
MicroRNAs (miRNAs) are a subset of small non-coding single-stranded RNA molecules involved in the regulation of post-transcriptional gene expression of a variety of transcript targets. Therefore altered miRNA expression may result in the dysregulation of key genes and biological pathways that has been reported with the onset and progression of neurodegenerative diseases, such as Amyotrophic lateral sclerosis (ALS). ALS is marked by a progressive degeneration of motor neurons (MNs) present in the spinal cord, brain stem and motor cortex. Although the pathomechanism underlying molecular interactions of ALS remains poorly understood, alterations in RNA metabolism, including dysregulation of miRNA expression in familial as well as sporadic forms are still scarcely studied. In this study, we performed combined transcriptomic data and miRNA profiling in MN samples of the same samples of iPSC-derived MNs from SOD1- and TARDBP (TDP-43 protein)-mutant-ALS patients and healthy controls. We report a global upregulation of mature miRNAs, and suggest that differentially expressed (DE) miRNAs have a significant impact on mRNA-level in SOD1-, but not in TARDBP-linked ALS. Furthermore, in SOD1-ALS we identified dysregulated miRNAs such as miR-124-3p, miR-19b-3p and miR-218 and their potential targets previously implicated in important functional process and pathogenic pathways underlying ALS. These miRNAs may play key roles in the neuronal development and cell survival related functions in SOD1-ALS. Altogether, we provide evidence of miRNA regulated genes expression mainly in SOD1 rather than TDP43-ALS.
Collapse
Affiliation(s)
- Banaja P Dash
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Gehlsheimer Str. 20, Rostock 18147, Germany
| | - Axel Freischmidt
- Department of Neurology, Ulm University, Albert-Einstein-Allee 11, Ulm 89081, Germany
| | - Jochen H Weishaupt
- Division of Neurodegeneration, Department of Neurology, Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Gehlsheimer Str. 20, Rostock 18147, Germany
- Center for Transdisciplinary Neurosciences Rostock, University Medical Center Rostock, Gehlsheimer Str. 20, Rostock 18147, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, Gehlsheimer Str. 20, Rostock 18147, Germany
| |
Collapse
|
7
|
Limone F, Mordes DA, Couto A, Joseph BJ, Mitchell JM, Therrien M, Ghosh SD, Meyer D, Zhang Y, Goldman M, Bortolin L, Cobos I, Stevens B, McCarroll SA, Kadiu I, Burberry A, Pietiläinen O, Eggan K. Single-nucleus sequencing reveals enriched expression of genetic risk factors in extratelencephalic neurons sensitive to degeneration in ALS. NATURE AGING 2024; 4:984-997. [PMID: 38907103 PMCID: PMC11257952 DOI: 10.1038/s43587-024-00640-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/01/2024] [Indexed: 06/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by a progressive loss of motor function linked to degenerating extratelencephalic neurons/Betz cells (ETNs). The reasons why these neurons are selectively affected remain unclear. Here, to understand the unique molecular properties that may sensitize ETNs to ALS, we performed RNA sequencing of 79,169 single nuclei from cortices of patients and controls. In both patients and unaffected individuals, we found significantly higher expression of ALS risk genes in THY1+ ETNs, regardless of diagnosis. In patients, this was accompanied by the induction of genes involved in protein homeostasis and stress responses that were significantly induced in a wide collection of ETNs. Examination of oligodendroglial and microglial nuclei revealed patient-specific downregulation of myelinating genes in oligodendrocytes and upregulation of an endolysosomal reactive state in microglia. Our findings suggest that selective vulnerability of extratelencephalic neurons is partly connected to their intrinsic molecular properties sensitizing them to genetics and mechanisms of degeneration.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
| | - Daniel A Mordes
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Brian J Joseph
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jana M Mitchell
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martine Therrien
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Sulagna Dia Ghosh
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Daniel Meyer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Yingying Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Melissa Goldman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Laura Bortolin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Inma Cobos
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Beth Stevens
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Steven A McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Irena Kadiu
- Neuroinflammation Focus Area, UCB Pharma, Braine-l'Alleud, Belgium
| | - Aaron Burberry
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Olli Pietiläinen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
8
|
Smith CEL, Laugel-Haushalter V, Hany U, Best S, Taylor RL, Poulter JA, Wortmann SB, Feichtinger RG, Mayr JA, Al Bahlani S, Nikolopoulos G, Rigby A, Black GC, Watson CM, Mansour S, Inglehearn CF, Mighell AJ, Bloch-Zupan A. Biallelic variants in Plexin B2 ( PLXNB2) cause amelogenesis imperfecta, hearing loss and intellectual disability. J Med Genet 2024; 61:689-698. [PMID: 38458752 PMCID: PMC11228227 DOI: 10.1136/jmg-2023-109728] [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: 11/02/2023] [Accepted: 02/22/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Plexins are large transmembrane receptors for the semaphorin family of signalling proteins. Semaphorin-plexin signalling controls cellular interactions that are critical during development as well as in adult life stages. Nine plexin genes have been identified in humans, but despite the apparent importance of plexins in development, only biallelic PLXND1 and PLXNA1 variants have so far been associated with Mendelian genetic disease. METHODS Eight individuals from six families presented with a recessively inherited variable clinical condition, with core features of amelogenesis imperfecta (AI) and sensorineural hearing loss (SNHL), with variable intellectual disability. Probands were investigated by exome or genome sequencing. Common variants and those unlikely to affect function were excluded. Variants consistent with autosomal recessive inheritance were prioritised. Variant segregation analysis was performed by Sanger sequencing. RNA expression analysis was conducted in C57Bl6 mice. RESULTS Rare biallelic pathogenic variants in plexin B2 (PLXNB2), a large transmembrane semaphorin receptor protein, were found to segregate with disease in all six families. The variants identified include missense, nonsense, splicing changes and a multiexon deletion. Plxnb2 expression was detected in differentiating ameloblasts. CONCLUSION We identify rare biallelic pathogenic variants in PLXNB2 as a cause of a new autosomal recessive, phenotypically diverse syndrome with AI and SNHL as core features. Intellectual disability, ocular disease, ear developmental abnormalities and lymphoedema were also present in multiple cases. The variable syndromic human phenotype overlaps with that seen in Plxnb2 knockout mice, and, together with the rarity of human PLXNB2 variants, may explain why pathogenic variants in PLXNB2 have not been reported previously.
Collapse
Affiliation(s)
- Claire E L Smith
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Virginie Laugel-Haushalter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS-UMR7104, Université de Strasbourg, Strasbourg, France
| | - Ummey Hany
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Sunayna Best
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Rachel L Taylor
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Evolution and Genomic Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
- EMQN CIC, Manchester, UK
| | - James A Poulter
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Saskia B Wortmann
- Department of Paediatrics, University Children's Hospital, Salzburger Landesklinken (SALK) and Paracelsus Medical University, Salzburg, Austria
- Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Rene G Feichtinger
- Department of Paediatrics, University Children's Hospital, Salzburger Landesklinken (SALK) and Paracelsus Medical University, Salzburg, Austria
| | - Johannes A Mayr
- Department of Paediatrics, University Children's Hospital, Salzburger Landesklinken (SALK) and Paracelsus Medical University, Salzburg, Austria
| | - Suhaila Al Bahlani
- Dental & OMFS Clinic, Al Nahdha Hospital, Government of Oman Ministry of Health, Muscat, Oman
| | | | - Alice Rigby
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
- School of Dentistry, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Graeme C Black
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Evolution and Genomic Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Christopher M Watson
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
- North East and Yorkshire Genomic Laboratory Hub, Central Lab, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sahar Mansour
- Lymphovascular Research Unit, Molecular and Clinical Sciences Research Institute, St George's Hospital, University of London, London, UK
- SW Thames Regional Centre for Genomics, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Chris F Inglehearn
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Alan J Mighell
- School of Dentistry, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Agnès Bloch-Zupan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS-UMR7104, Université de Strasbourg, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
- Centre de référence des maladies rares orales et dentaires O-Rares, Filière Santé Maladies rares TETE COU, European Reference Network CRANIO, Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpital Civil, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France
| |
Collapse
|
9
|
Wang HLV, Xiang JF, Yuan C, Veire AM, Gendron TF, Murray ME, Tansey MG, Hu J, Gearing M, Glass JD, Jin P, Corces VG, McEachin ZT. pTDP-43 levels correlate with cell type specific molecular alterations in the prefrontal cortex of C9orf72 ALS/FTD patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.12.523820. [PMID: 36711601 PMCID: PMC9882184 DOI: 10.1101/2023.01.12.523820] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Repeat expansions in the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis and familial frontotemporal dementia (ALS/FTD). To identify molecular defects that take place in the dorsolateral frontal cortex of patients with C9orf72 ALS/FTD, we compared healthy controls with C9orf72 ALS/FTD donor samples staged based on the levels of cortical phosphorylated TAR DNA binding protein (pTDP-43), a neuropathological hallmark of disease progression. We identified distinct molecular changes in different cell types that take place during FTD development. Loss of neurosurveillance microglia and activation of the complement cascade take place early, when pTDP-43 aggregates are absent or very low, and become more pronounced in late stages, suggesting an initial involvement of microglia in disease progression. Reduction of layer 2-3 cortical projection neurons with high expression of CUX2/LAMP5 also occurs early, and the reduction becomes more pronounced as pTDP-43 accumulates. Several unique features were observed only in samples with high levels of pTDP-43, including global alteration of chromatin accessibility in oligodendrocytes, microglia, and astrocytes; higher ratios of premature oligodendrocytes; increased levels of the noncoding RNA NEAT1 in astrocytes and neurons, and higher amount of phosphorylated ribosomal protein S6. Our findings reveal previously unknown progressive functional changes in major cell types found in the frontal cortex of C9orf72 ALS/FTD patients that shed light on the mechanisms underlying the pathology of this disease.
Collapse
Affiliation(s)
- Hsiao-Lin V. Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Jian-Feng Xiang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Chenyang Yuan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Austin M. Veire
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | | | | | - Malú G. Tansey
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32607
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32607
| | - Jian Hu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Marla Gearing
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Jonathan D. Glass
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Victor G. Corces
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Zachary T. McEachin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
10
|
Cauchi RJ. SCFD1 in amyotrophic lateral sclerosis: reconciling a genetic association with in vivo functional analysis. Neural Regen Res 2024; 19:1201-1202. [PMID: 37905864 PMCID: PMC11467948 DOI: 10.4103/1673-5374.386411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Ruben J. Cauchi
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building; Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| |
Collapse
|
11
|
Odierna GL, Vucic S, Dyer M, Dickson T, Woodhouse A, Blizzard C. How do we get from hyperexcitability to excitotoxicity in amyotrophic lateral sclerosis? Brain 2024; 147:1610-1621. [PMID: 38408864 PMCID: PMC11068114 DOI: 10.1093/brain/awae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 02/28/2024] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease that, at present, has no effective cure. Evidence of increased circulating glutamate and hyperexcitability of the motor cortex in patients with amyotrophic lateral sclerosis have provided an empirical support base for the 'dying forward' excitotoxicity hypothesis. The hypothesis postulates that increased activation of upper motor neurons spreads pathology to lower motor neurons in the spinal cord in the form of excessive glutamate release, which triggers excitotoxic processes. Many clinical trials have focused on therapies that target excitotoxicity via dampening neuronal activation, but not all are effective. As such, there is a growing tension between the rising tide of evidence for the 'dying forward' excitotoxicity hypothesis and the failure of therapies that target neuronal activation. One possible solution to these contradictory outcomes is that our interpretation of the current evidence requires revision in the context of appreciating the complexity of the nervous system and the limitations of the neurobiological assays we use to study it. In this review we provide an evaluation of evidence relevant to the 'dying forward' excitotoxicity hypothesis and by doing so, identify key gaps in our knowledge that need to be addressed. We hope to provide a road map from hyperexcitability to excitotoxicity so that we can better develop therapies for patients suffering from amyotrophic lateral sclerosis. We conclude that studies of upper motor neuron activity and their synaptic output will play a decisive role in the future of amyotrophic lateral sclerosis therapy.
Collapse
Affiliation(s)
- G Lorenzo Odierna
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney 2050, Australia
| | - Marcus Dyer
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- Department of Pharmaceutical and Pharmacological Sciences, Center for Neurosciences, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Tracey Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Adele Woodhouse
- The Wicking Dementia Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Catherine Blizzard
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
12
|
Kim SH, Oh KW, Noh MY, Kwon MS. Optimal Therapeutic Strategy of Bone Marrow-Originated Autologous Mesenchymal Stromal/Stem Cells for ALS. Stem Cells Transl Med 2024; 13:309-316. [PMID: 38244235 PMCID: PMC11016834 DOI: 10.1093/stcltm/szad095] [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: 09/30/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by selective and progressive neurodegenerative changes in motor neural networks. Given the system complexity, including anatomically distributed sites of degeneration from the motor cortex to the spinal cord and chronic pro-inflammatory conditions, a cell-based therapeutic strategy could be an alternative approach to treating ALS. Lessons from previous mesenchymal stromal/stem cell (MSC) trials in ALS realized the importance of 3 aspects in current and future MSC therapy, including the preparation of MSCs, administration routes and methods, and recipient-related factors. This review briefly describes the current status and future prerequisites for an optimal strategy using bone-marrow-originated MSCs to treat ALS. We suggest mandatory factors in the optimized therapeutic strategy focused on advanced therapy medicinal products produced according to Good Manufacturing Practice, an optimal administration method, the selection of proper patients, and the importance of biomarkers.
Collapse
Affiliation(s)
- Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Ki-Wook Oh
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Min-Young Noh
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute of Basic Medical Science, School of Medicine, CHA University, CHA Bio Complex, Gyeonggi-do, Republic of Korea
| |
Collapse
|
13
|
He D, Liu Y, Dong S, Shen D, Yang X, Hao M, Yin X, He X, Li Y, Wang Y, Liu M, Wang J, Chen X, Cui L. The prognostic value of systematic genetic screening in amyotrophic lateral sclerosis patients. J Neurol 2024; 271:1385-1396. [PMID: 37980296 DOI: 10.1007/s00415-023-12079-1] [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: 09/06/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/20/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with complex genetic architecture. Emerging evidence has indicated comorbidity between ALS and autoimmune conditions, suggesting a potential shared genetic basis. The objective of this study is to assess the prognostic value of systematic screening for rare deleterious mutations in genes associated with ALS and aberrant inflammatory responses. METHODS A discovery cohort of 494 patients and a validation cohort of 69 patients were analyzed in this study, with population-matched healthy subjects (n = 4961) served as controls. Whole exome sequencing (WES) was performed to identify rare deleterious variants in 50 ALS genes and 1177 genes associated with abnormal inflammatory responses. Genotype-phenotype correlation was assessed, and an integrative prognostic model incorporating genetic and clinical factors was constructed. RESULTS In the discovery cohort, 8.1% of patients carried confirmed ALS variants, and an additional 15.2% of patients carried novel ALS variants. Gene burden analysis revealed 303 immune-implicated genes with enriched rare variants, and 13.4% of patients harbored rare deleterious variants in these genes. Patients with ALS variants exhibited a more rapid disease progression (HR 2.87 [95% CI 2.03-4.07], p < 0.0001), while no significant effect was observed for immune-implicated variants. The nomogram model incorporating genetic and clinical information demonstrated improved accuracy in predicting disease outcomes (C-index, 0.749). CONCLUSION Our findings enhance the comprehension of the genetic basis of ALS within the Chinese population. It also appears that rare deleterious mutations occurring in immune-implicated genes exert minimal influence on the clinical trajectories of ALS patients.
Collapse
Affiliation(s)
- Di He
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Yining Liu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Siqi Dong
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Dongchao Shen
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Xunzhe Yang
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Meng Hao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Xianhong Yin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Xinyi He
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yi Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yi Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Mingsheng Liu
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China.
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China.
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China.
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China.
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China.
| |
Collapse
|
14
|
Monov D, Molodozhnikova N. Biochemical parameters as a tool to assess the nutritional status of patients with amyotrophic lateral sclerosis. Front Neurol 2024; 14:1258224. [PMID: 38313408 PMCID: PMC10836144 DOI: 10.3389/fneur.2023.1258224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/27/2023] [Indexed: 02/06/2024] Open
Abstract
Background The research aimed to analyze blood biochemical parameters in patients with amyotrophic lateral sclerosis and to determine whether they can be used to assess their nutritional status. Methods The study included 45 patients diagnosed with amyotrophic lateral sclerosis (ALS): 28 (62.2%) were men and 17 (37.8%) were women. The mean age of the study participants was 50.69 ± 7.24 years. The control group consisted of 30 practically healthy individuals. Results Compared with practically healthy individuals, patients with ALS had significantly lower blood parameters, including total lymphocyte count (1.49 ± 0.11 vs. 2.86 ± 0.25, p < 0.05), total protein (60.55 ± 2.38 vs. 77.80 ± 4.41, p < 0.05), albumin (33.70 ± 2.03 vs. 46.49 ± 3.22, p < 0.05), urea (3.09 ± 0.36 vs. 5.37 ± 0.50, p < 0.05), creatinine (51.28 ± 4.42 vs. 70.91 ± 5.13, p < 0.05), and transferrin (1.84 ± 0.12 vs. 2.32 ± 0.10, p < 0.05). These parameters correspond to first-degree malnutrition. There were direct correlations between anthropometric and biochemical parameters in the ALS group. BMI correlated with the blood levels of total protein (r = 0.22, p < 0.05), albumin (r = 0.27, p < 0.05), urea (r = 0.33, p < 0.05), creatinine (r = 0.30, p < 0.05), transferrin (r = 0.18, p < 0.05), and total lymphocyte count (r = 0.20, p < 0.05). PNI correlated with the blood levels of total protein (r = 0.53, p < 0.05), albumin (r = 0.87, p < 0.05), total cholesterol (r = 0.34, p < 0.05), transferrin (r = 0.40, p < 0.05), total lymphocyte count (r = 0.79, p < 0.05), urea (r = 0, 37, p < 0.05), and creatinine (r = 0.32, p < 0.05). Conclusion The study presents compelling evidence supporting the utilization of biochemical parameters, including total protein, albumin, urea, creatinine, transferrin, and total lymphocyte count, for potentially evaluating the nutritional status of individuals diagnosed with ALS.
Collapse
Affiliation(s)
- Dimitar Monov
- Department of Anesthesiology and Intensive Care, Medical University of Sofia, Sofia, Bulgaria
| | - Natalia Molodozhnikova
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
15
|
Zhu Y, Burg T, Neyrinck K, Vervliet T, Nami F, Vervoort E, Ahuja K, Sassano ML, Chai YC, Tharkeshwar AK, De Smedt J, Hu H, Bultynck G, Agostinis P, Swinnen JV, Van Den Bosch L, da Costa RFM, Verfaillie C. Disruption of MAM integrity in mutant FUS oligodendroglial progenitors from hiPSCs. Acta Neuropathol 2024; 147:6. [PMID: 38170217 PMCID: PMC10764485 DOI: 10.1007/s00401-023-02666-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disorder, characterized by selective loss of motor neurons (MNs). A number of causative genetic mutations underlie the disease, including mutations in the fused in sarcoma (FUS) gene, which can lead to both juvenile and late-onset ALS. Although ALS results from MN death, there is evidence that dysfunctional glial cells, including oligodendroglia, contribute to neurodegeneration. Here, we used human induced pluripotent stem cells (hiPSCs) with a R521H or a P525L mutation in FUS and their isogenic controls to generate oligodendrocyte progenitor cells (OPCs) by inducing SOX10 expression from a TET-On SOX10 cassette. Mutant and control iPSCs differentiated efficiently into OPCs. RNA sequencing identified a myelin sheath-related phenotype in mutant OPCs. Lipidomic studies demonstrated defects in myelin-related lipids, with a reduction of glycerophospholipids in mutant OPCs. Interestingly, FUSR521H OPCs displayed a decrease in the phosphatidylcholine/phosphatidylethanolamine ratio, known to be associated with maintaining membrane integrity. A proximity ligation assay further indicated that mitochondria-associated endoplasmic reticulum membranes (MAM) were diminished in both mutant FUS OPCs. Moreover, both mutant FUS OPCs displayed increased susceptibility to ER stress when exposed to thapsigargin, and exhibited impaired mitochondrial respiration and reduced Ca2+ signaling from ER Ca2+ stores. Taken together, these results demonstrate a pathological role of mutant FUS in OPCs, causing defects in lipid metabolism associated with MAM disruption manifested by impaired mitochondrial metabolism with increased susceptibility to ER stress and with suppressed physiological Ca2+ signaling. As such, further exploration of the role of oligodendrocyte dysfunction in the demise of MNs is crucial and will provide new insights into the complex cellular mechanisms underlying ALS.
Collapse
Affiliation(s)
- Yingli Zhu
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium.
| | - Thibaut Burg
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | - Katrien Neyrinck
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Fatemeharefeh Nami
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Ellen Vervoort
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Karan Ahuja
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
- Animal Physiology and Neurobiology Section, Department of Biology, Neural Circuit Development and Regeneration Research Group, 3000, Leuven, Belgium
| | - Maria Livia Sassano
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Yoke Chin Chai
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | - Jonathan De Smedt
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Haibo Hu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, 3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | | | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| |
Collapse
|
16
|
Holtman IR, Glass CK, Nott A. Interpretation of Neurodegenerative GWAS Risk Alleles in Microglia and their Interplay with Other Cell Types. ADVANCES IN NEUROBIOLOGY 2024; 37:531-544. [PMID: 39207711 DOI: 10.1007/978-3-031-55529-9_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia have been implicated in numerous neurodegenerative and neuroinflammatory disorders; however, the causal contribution of this immune cell type is frequently debated. Genetic studies offer a unique vantage point in that they infer causality over a secondary consequence. Genome-wide association studies (GWASs) have identified hundreds of loci in the genome that are associated with susceptibility to neurodegenerative disorders. GWAS studies implicate microglia in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and to a lesser degree suggest a role for microglia in vascular dementia (VaD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS), and other neurodegenerative and neuropsychiatric disorders. The contribution and function of GWAS risk loci on disease progression is an ongoing field of study, in which large genomic datasets, and an extensive framework of computational tools, have proven to be crucial. Several GWAS risk loci are shared between disorders, pointing towards common pleiotropic mechanisms. In this chapter, we introduce key concepts in GWAS and post-GWAS interpretation of neurodegenerative disorders, with a focus on GWAS risk genes implicated in microglia, their interplay with other cell types and shared convergence of GWAS risk loci on microglia.
Collapse
Affiliation(s)
- Inge R Holtman
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
- Department of Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
| | - Alexi Nott
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| |
Collapse
|
17
|
Paris A, Lakatos A. Cell and gene therapy for amyotrophic lateral sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:217-241. [PMID: 39341656 DOI: 10.1016/b978-0-323-90120-8.00017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disorder with rapidly progressive skeletal muscle weakness, which can also cause a variable cognitive deficit. Genetic causes are only identified in approximately 10% of all cases, with complex genotype-phenotype associations, making it challenging to identify treatment targets. What further hampers therapeutic development is a broad heterogeneity in mechanisms, possible targets, and disturbances across various cell types, aside from the cortical and spinal motor neurons that lie at the heart of the pathology of ALS. Over the last decade, significant progress in biotechnologic techniques, cell and ribonucleic acid (RNA) engineering, animal models, and patient-specific human stem cell and organoid models have accelerated both mechanistic and therapeutic discoveries. The growing number of clinical trials mirrors this. This chapter reviews the current state of human preclinical models supporting trial strategies as well as recent clinical cell and gene therapy approaches.
Collapse
Affiliation(s)
- Alvar Paris
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom; Department of Neurology, Cambridge University Hospitals NHS Trust, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - András Lakatos
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom; Department of Neurology, Cambridge University Hospitals NHS Trust, Addenbrooke's Hospital, Cambridge, United Kingdom.
| |
Collapse
|
18
|
Tang L, Tang X, Zhao Q, Li Y, Bu Y, Liu Z, Li J, Guo J, Shen L, Jiang H, Tang B, Xu R, Cao W, Yuan Y, Wang J. Mutation and clinical analysis of the CLCC1 gene in amyotrophic lateral sclerosis patients from Central South China. Ann Clin Transl Neurol 2024; 11:79-88. [PMID: 37916886 PMCID: PMC10791024 DOI: 10.1002/acn3.51934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 11/03/2023] Open
Abstract
INTRODUCTION Recently, chloride channel CLIC-like 1 (CLCC1) was reported to be a novel ALS-related gene. We aimed to screen CLCC1 variants in our ALS cohort and further explore the genotype-phenotype correlation of CLCC1-related ALS. METHODS We screened rare damaging variants in CLCC1 from our cohorts of 1005 ALS patients and 1224 healthy controls with whole-exome sequencing in Central South China. Fisher's exact test was conducted for association analysis at the entire gene level and single variant level. RESULTS In total, four heterozygous missense variants in CLCC1 were identified from four unrelated sporadic ALS patients and predicted to be putative pathogenic by in silico tools and protein model prediction, accounting for 0.40% of all patients (4/1005). The four variants were c.A275C (p.Q92P), c.G1139A (p.R380K), c.C1244T (p.T415M), and c.G1328A (p.R443Q), respectively, which had not been reported in ALS patients previously. Three of four variants were located in exon 10. Patients harboring CLCC1 variants seemed to share a group of similar clinical features, including earlier age at onset, rapid progression, spinal onset, and vulnerable cognitive status. Statistically, we did not find CLCC1 to be associated with the risk of ALS at the entire gene level or single variant level. CONCLUSION Our findings further expanded the genetic and clinical spectrum of CLCC1-related ALS and provided more genetic evidence for anion channel involvement in the pathogenesis of ALS, but further investigations are needed to verify our findings.
Collapse
Affiliation(s)
- Linxin Tang
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
| | - Xuxiong Tang
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
| | - Qianqian Zhao
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
| | - Yongchao Li
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
| | - Yue Bu
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
| | - Zhen Liu
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
| | - Jinchen Li
- National Clinical Research Center for Geriatric Diseases, Xiangya HospitalCentral South UniversityChangshaP. R. China
- Center for Medical Genetics, School of Life SciencesCentral South UniversityChangshaP. R. China
- Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaP. R. China
| | - Jifeng Guo
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
- National Clinical Research Center for Geriatric Diseases, Xiangya HospitalCentral South UniversityChangshaP. R. China
- Center for Medical Genetics, School of Life SciencesCentral South UniversityChangshaP. R. China
- Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaP. R. China
- Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaP. R. China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaP. R. China
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative DiseasesChangshaP. R. China
| | - Lu Shen
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
- National Clinical Research Center for Geriatric Diseases, Xiangya HospitalCentral South UniversityChangshaP. R. China
- Center for Medical Genetics, School of Life SciencesCentral South UniversityChangshaP. R. China
- Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaP. R. China
- Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaP. R. China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaP. R. China
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative DiseasesChangshaP. R. China
| | - Hong Jiang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
- National Clinical Research Center for Geriatric Diseases, Xiangya HospitalCentral South UniversityChangshaP. R. China
- Center for Medical Genetics, School of Life SciencesCentral South UniversityChangshaP. R. China
- Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaP. R. China
- Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaP. R. China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaP. R. China
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative DiseasesChangshaP. R. China
| | - Beisha Tang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
- National Clinical Research Center for Geriatric Diseases, Xiangya HospitalCentral South UniversityChangshaP. R. China
- Center for Medical Genetics, School of Life SciencesCentral South UniversityChangshaP. R. China
- Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaP. R. China
- Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaP. R. China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaP. R. China
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative DiseasesChangshaP. R. China
| | - Renshi Xu
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical CollegeFirst Affiliated Hospital of Nanchang Medical CollegeNanchangP. R. China
| | - Wenfeng Cao
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical CollegeFirst Affiliated Hospital of Nanchang Medical CollegeNanchangP. R. China
| | - Yanchun Yuan
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
| | - Junling Wang
- Department of Neurology, Xiangya HospitalCentral South University, Jiangxi Hospital, National Regional Center for Neurological DiseasesNanchangP. R. China
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaP. R. China
- National Clinical Research Center for Geriatric Diseases, Xiangya HospitalCentral South UniversityChangshaP. R. China
- Center for Medical Genetics, School of Life SciencesCentral South UniversityChangshaP. R. China
- Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaP. R. China
- Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaP. R. China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaP. R. China
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative DiseasesChangshaP. R. China
| |
Collapse
|
19
|
Festa LK, Grinspan JB, Jordan-Sciutto KL. White matter injury across neurodegenerative disease. Trends Neurosci 2024; 47:47-57. [PMID: 38052682 PMCID: PMC10842057 DOI: 10.1016/j.tins.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/16/2023] [Accepted: 11/11/2023] [Indexed: 12/07/2023]
Abstract
Oligodendrocytes (OLs), the myelin-generating cells of the central nervous system (CNS), are active players in shaping neuronal circuitry and function. It has become increasingly apparent that injury to cells within the OL lineage plays a central role in neurodegeneration. In this review, we focus primarily on three degenerative disorders in which white matter loss is well documented: Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). We discuss clinical data implicating white matter injury as a key feature of these disorders, as well as shared and divergent phenotypes between them. We examine the cellular and molecular mechanisms underlying the alterations to OLs, including chronic neuroinflammation, aggregation of proteins, lipid dysregulation, and organellar stress. Last, we highlight prospects for therapeutic intervention targeting the OL lineage to restore function.
Collapse
Affiliation(s)
- Lindsay K Festa
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Judith B Grinspan
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kelly L Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Sattler R, Traynor BJ, Robertson J, Van Den Bosch L, Barmada SJ, Svendsen CN, Disney MD, Gendron TF, Wong PC, Turner MR, Boxer A, Babu S, Benatar M, Kurnellas M, Rohrer JD, Donnelly CJ, Bustos LM, Van Keuren-Jensen K, Dacks PA, Sabbagh MN. Roadmap for C9ORF72 in Frontotemporal Dementia and Amyotrophic Lateral Sclerosis: Report on the C9ORF72 FTD/ALS Summit. Neurol Ther 2023; 12:1821-1843. [PMID: 37847372 PMCID: PMC10630271 DOI: 10.1007/s40120-023-00548-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/14/2023] [Indexed: 10/18/2023] Open
Abstract
A summit held March 2023 in Scottsdale, Arizona (USA) focused on the intronic hexanucleotide expansion in the C9ORF72 gene and its relevance in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS; C9ORF72-FTD/ALS). The goal of this summit was to connect basic scientists, clinical researchers, drug developers, and individuals affected by C9ORF72-FTD/ALS to evaluate how collaborative efforts across the FTD-ALS disease spectrum might break down existing disease silos. Presentations and discussions covered recent discoveries in C9ORF72-FTD/ALS disease mechanisms, availability of disease biomarkers and recent advances in therapeutic development, and clinical trial design for prevention and treatment for individuals affected by C9ORF72-FTD/ALS and asymptomatic pathological expansion carriers. The C9ORF72-associated hexanucleotide repeat expansion is an important locus for both ALS and FTD. C9ORF72-FTD/ALS may be characterized by loss of function of the C9ORF72 protein and toxic gain of functions caused by both dipeptide repeat (DPR) proteins and hexanucleotide repeat RNA. C9ORF72-FTD/ALS therapeutic strategies discussed at the summit included the use of antisense oligonucleotides, adeno-associated virus (AAV)-mediated gene silencing and gene delivery, and engineered small molecules targeting RNA structures associated with the C9ORF72 expansion. Neurofilament light chain, DPR proteins, and transactive response (TAR) DNA-binding protein 43 (TDP-43)-associated molecular changes were presented as biomarker candidates. Similarly, brain imaging modalities (i.e., magnetic resonance imaging [MRI] and positron emission tomography [PET]) measuring structural, functional, and metabolic changes were discussed as important tools to monitor individuals affected with C9ORF72-FTD/ALS, at both pre-symptomatic and symptomatic disease stages. Finally, summit attendees evaluated current clinical trial designs available for FTD or ALS patients and concluded that therapeutics relevant to FTD/ALS patients, such as those specifically targeting C9ORF72, may need to be tested with composite endpoints covering clinical symptoms of both FTD and ALS. The latter will require novel clinical trial designs to be inclusive of all patient subgroups spanning the FTD/ALS spectrum.
Collapse
Affiliation(s)
- Rita Sattler
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ludo Van Den Bosch
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology and KU Leuven, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
| | - Sami J Barmada
- Department of Neurology, Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Matthew D Disney
- Department of Chemistry, The Herbert Wertheim UF-Scripps Institute for Biomedical Research and Innovation, The Scripps Research Institute, Jupiter, FL, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Philip C Wong
- Departments of Pathology and Neuroscience, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adam Boxer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of San Francisco, San Francisco, CA, USA
| | - Suma Babu
- Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital-Harvard Medical School, Boston, MA, USA
| | - Michael Benatar
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33129, USA
| | | | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher J Donnelly
- LiveLikeLou Center for ALS Research, Brain Institute, University of Pittsburgh, Pittsburgh, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lynette M Bustos
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA
| | | | - Penny A Dacks
- The Association for Frontotemporal Degeneration and FTD Disorders Registry, King of Prussia, PA, USA
| | - Marwan N Sabbagh
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| |
Collapse
|
21
|
Rizzuti M, Sali L, Melzi V, Scarcella S, Costamagna G, Ottoboni L, Quetti L, Brambilla L, Papadimitriou D, Verde F, Ratti A, Ticozzi N, Comi GP, Corti S, Gagliardi D. Genomic and transcriptomic advances in amyotrophic lateral sclerosis. Ageing Res Rev 2023; 92:102126. [PMID: 37972860 DOI: 10.1016/j.arr.2023.102126] [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: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder and the most common motor neuron disease. ALS shows substantial clinical and molecular heterogeneity. In vitro and in vivo models coupled with multiomic techniques have provided important contributions to unraveling the pathomechanisms underlying ALS. To date, despite promising results and accumulating knowledge, an effective treatment is still lacking. Here, we provide an overview of the literature on the use of genomics, epigenomics, transcriptomics and microRNAs to deeply investigate the molecular mechanisms developing and sustaining ALS. We report the most relevant genes implicated in ALS pathogenesis, discussing the use of different high-throughput sequencing techniques and the role of epigenomic modifications. Furthermore, we present transcriptomic studies discussing the most recent advances, from microarrays to bulk and single-cell RNA sequencing. Finally, we discuss the use of microRNAs as potential biomarkers and promising tools for molecular intervention. The integration of data from multiple omic approaches may provide new insights into pathogenic pathways in ALS by shedding light on diagnostic and prognostic biomarkers, helping to stratify patients into clinically relevant subgroups, revealing novel therapeutic targets and supporting the development of new effective therapies.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simone Scarcella
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Gianluca Costamagna
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Linda Ottoboni
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Federico Verde
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Nicola Ticozzi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| | - Delia Gagliardi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
22
|
Lee A, Henderson R, Arachchige BJ, Robertson T, McCombe PA. Proteomic investigation of ALS motor cortex identifies known and novel pathogenetic mechanisms. J Neurol Sci 2023; 452:120753. [PMID: 37542825 DOI: 10.1016/j.jns.2023.120753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/30/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
The key pathological feature in ALS is death of motor neurones from the brain and spinal cord, but the molecular mechanisms underlying this degeneration remain unknown. Quantifying the motor cortex proteome in autopsy brain and comparing tissues from ALS cases and non-ALS controls is critical to understanding these mechanisms. We used Sequential Window Acquisition of All Theoretical Mass Spectra (SWATH-MS) to characterize the proteomes of the motor cortex from ALS cases (n = 8) and control subjects (n = 8). A total of 1427 proteins were identified at a critical local false discovery rate < 5%; 187 of these exhibited significant expression differences between ALS cases and controls. Of these, 91 proteins were significantly upregulated and 96 proteins were significantly downregulated. Bioinformatics analysis revealed that these proteins are involved in molecular transport, protein trafficking, free radical scavenging, lipid metabolism, cell death and survival, nucleic acid metabolism, inflammatory response or amino acid metabolism and carbohydrate metabolism. Differentially expressed proteins were subjected to pathway analysis. This revealed abnormalities in pathways involving mitochondrial function, sirtuin signaling, oxidative phosphorylation, glycolysis, phagosome maturation, SNARE signaling, redox regulation and several others. Core analysis revealed mitochondrial dysfunction to be the top canonical pathway. The top-enriched networks involved JNK activation and inhibition of AKT signaling, suggesting that disruption of these signaling pathways could lead to demise of motor neurons in the ALS motor cortex.
Collapse
Affiliation(s)
- Aven Lee
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Robert Henderson
- Department of Neurology, Royal Brisbane & Women's Hospital (RBWH), Brisbane, QLD 4029, Australia
| | - Buddhika Jayakody Arachchige
- Mass Spectrometry Facility, Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Thomas Robertson
- Pathology, Royal Brisbane & Women's Hospital, Brisbane, QLD 4029, Australia; School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Pamela Ann McCombe
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; Wesley Medical Research, The Wesley Hospital, Auchenflower, QLD 4066, Australia.
| |
Collapse
|
23
|
Akçimen F, Lopez ER, Landers JE, Nath A, Chiò A, Chia R, Traynor BJ. Amyotrophic lateral sclerosis: translating genetic discoveries into therapies. Nat Rev Genet 2023; 24:642-658. [PMID: 37024676 PMCID: PMC10611979 DOI: 10.1038/s41576-023-00592-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2023] [Indexed: 04/08/2023]
Abstract
Recent advances in sequencing technologies and collaborative efforts have led to substantial progress in identifying the genetic causes of amyotrophic lateral sclerosis (ALS). This momentum has, in turn, fostered the development of putative molecular therapies. In this Review, we outline the current genetic knowledge, emphasizing recent discoveries and emerging concepts such as the implication of distinct types of mutation, variability in mutated genes in diverse genetic ancestries and gene-environment interactions. We also propose a high-level model to synthesize the interdependent effects of genetics, environmental and lifestyle factors, and ageing into a unified theory of ALS. Furthermore, we summarize the current status of therapies developed on the basis of genetic knowledge established for ALS over the past 30 years, and we discuss how developing treatments for ALS will advance our understanding of targeting other neurological diseases.
Collapse
Affiliation(s)
- Fulya Akçimen
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Elia R Lopez
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Adriano Chiò
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy
- Azienda Ospedaliero Universitaria Citta' della Salute e della Scienza, Turin, Italy
| | - Ruth Chia
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Bryan J Traynor
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA.
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA.
| |
Collapse
|
24
|
Dou J, Bakulski K, Guo K, Hur J, Zhao L, Saez-Atienzar S, Stark A, Chia R, García-Redondo A, Rojas-Garcia R, Vázquez Costa JF, Fernandez Santiago R, Bandres-Ciga S, Gómez-Garre P, Periñán MT, Mir P, Pérez-Tur J, Cardona F, Menendez-Gonzalez M, Riancho J, Borrego-Hernández D, Galán-Dávila L, Infante Ceberio J, Pastor P, Paradas C, Dols-Icardo O, Traynor BJ, Feldman EL, Goutman SA. Cumulative Genetic Score and C9orf72 Repeat Status Independently Contribute to Amyotrophic Lateral Sclerosis Risk in 2 Case-Control Studies. Neurol Genet 2023; 9:e200079. [PMID: 37293291 PMCID: PMC10245939 DOI: 10.1212/nxg.0000000000200079] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/06/2023] [Indexed: 06/10/2023]
Abstract
Background and Objectives Most patients with amyotrophic lateral sclerosis (ALS) lack a monogenic mutation. This study evaluates ALS cumulative genetic risk in an independent Michigan and Spanish replication cohort using polygenic scores. Methods Participant samples from University of Michigan were genotyped and assayed for the chromosome 9 open reading frame 72 hexanucleotide expansion. Final cohort size was 219 ALS and 223 healthy controls after genotyping and participant filtering. Polygenic scores excluding the C9 region were generated using an independent ALS genome-wide association study (20,806 cases, 59,804 controls). Adjusted logistic regression and receiver operating characteristic curves evaluated the association and classification between polygenic scores and ALS status, respectively. Population attributable fractions and pathway analyses were conducted. An independent Spanish study sample (548 cases, 2,756 controls) was used for replication. Results Polygenic scores constructed from 275 single-nucleotide variation (SNV) had the best model fit in the Michigan cohort. An SD increase in ALS polygenic score associated with 1.28 (95% CI 1.04-1.57) times higher odds of ALS with area under the curve of 0.663 vs a model without the ALS polygenic score (p value = 1 × 10-6). The population attributable fraction of the highest 20th percentile of ALS polygenic scores, relative to the lowest 80th percentile, was 4.1% of ALS cases. Genes annotated to this polygenic score enriched for important ALS pathomechanisms. Meta-analysis with the Spanish study, using a harmonized 132 single nucleotide variation polygenic score, yielded similar logistic regression findings (odds ratio: 1.13, 95% CI 1.04-1.23). Discussion ALS polygenic scores can account for cumulative genetic risk in populations and reflect disease-relevant pathways. If further validated, this polygenic score will inform future ALS risk models.
Collapse
Affiliation(s)
- John Dou
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Kelly Bakulski
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Kai Guo
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Junguk Hur
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Lili Zhao
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Sara Saez-Atienzar
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Ali Stark
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Ruth Chia
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Alberto García-Redondo
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Ricardo Rojas-Garcia
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Juan Francisco Vázquez Costa
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Ruben Fernandez Santiago
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Sara Bandres-Ciga
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Pilar Gómez-Garre
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Maria Teresa Periñán
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Pablo Mir
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Jordi Pérez-Tur
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Fernando Cardona
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Manuel Menendez-Gonzalez
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Javier Riancho
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Daniel Borrego-Hernández
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Lucia Galán-Dávila
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Jon Infante Ceberio
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Pau Pastor
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Carmen Paradas
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Oriol Dols-Icardo
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Bryan J Traynor
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Eva L Feldman
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| | - Stephen A Goutman
- From the Department of Epidemiology (J.D., K.B.), School of Public Health, Department of Neurology (K.G., E.L.F., S.A.G.), NeuroNetwork for Emerging Therapies (K.G., E.L.F., S.A.G.), University of Michigan, Ann Arbor; Department of Biomedical Sciences (J.H.), University of North Dakota, Grand Forks; Department of Biostatistics (L.Z.), School of Public Health, University of Michigan, Ann Arbor; Neuromuscular Diseases Research Section (S.S.-A., A.S., R.C., B.J.T.), Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD; ALS Unit (A.G.-R., D.B.-H.), Instituto de Investigación Sanitaria "i + 12" del Hospital Universitario 12 de Octubre de Madrid, SERMAS, CIBERER (A.G.-R., R.R.-G., J.F.V.C., D.B.-H.), Center for Networked Biomedical Research into Rare Diseases, Madrid; Neuromuscular Disorders Unit (R.R.-G.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona; Neuromuscular Unit (J.F.V.C.), Hospital Universitario y Politécnico la Fe, IIS La Fe; Department of Medicine (J.F.V.C.), Universitat de València; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) (R.F.S., P.G.-G., M.T.P., P.M., J.P.-T., F.C., O.D.-I.), Madrid; Lab of Parkinson's disease and Other Neurodegenerative Movement Disorders (R.F.S.), IDIBAPS-Institut d'Investigacions Biomèdiques, Barcelona; Unitat de Parkinson i Trastorns del Moviment, Servicio de Neurologia (R.F.S.), Hospital Clínic de Barcelona and Institut de Neurociencies de la Universitat de Barcelona (Maria de Maetzu Center), Catalonia, Spain; Center for Alzheimer's and Related Dementias (S.B.-C.), National Institute on Aging, Bethesda, MD; Unidad de Trastornos del Movimiento (P.G.-G., M.T.P., P.M.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC; Departamento de Medicina (P.M.), Universidad de Sevilla; Neurology and Molecular Genetics Mixed Investigation Unit (J.P.-T., F.C.), Instituto de Investigación Sanitaria La Fe, Molecular Genetics Unit (J.P.-T., F.C.), Institut de Biomedicina de València-CSIC; Department of Medicine (M.M.-G.), Universidad de Oviedo; Department of Neurology (M.M.-G.), Hospital Universitario Central de Asturias; Instituto de Investigación Sanitaria del Principado de Asturias (M.M.-G.), Oviedo, Spain; Service of Neurology (J.R.), Hospital Sierrallana, IDIVAL University of Cantabria, Torrelavega; Instituto de Investigación Marqués de Valdecilla (J.R., J.I.C.), Santander; Department of Neurology (L.G.-D.), ALS Unit, Hospital Clínico Universitario "San Carlos," Madrid; Unit of Neurodegenerative Diseases (P.P.), Department of Neurology, University Hospital Germans Trias I Pujol; Neurosciences (P.P.), The Germans Trias i Pujol Research Institute (IGTP) Badalona; Department of Neurology (C.P.), Hospital Universitario Virgen del Rocio, Sevilla; and Memory Unit (O.D.-I.), Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Spain
| |
Collapse
|
25
|
Szebényi K, Barrio-Hernandez I, Gibbons GM, Biasetti L, Troakes C, Beltrao P, Lakatos A. A human proteogenomic-cellular framework identifies KIF5A as a modulator of astrocyte process integrity with relevance to ALS. Commun Biol 2023; 6:678. [PMID: 37386082 PMCID: PMC10310856 DOI: 10.1038/s42003-023-05041-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 06/13/2023] [Indexed: 07/01/2023] Open
Abstract
Genome-wide association studies identified several disease-causing mutations in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). However, the contribution of genetic variants to pathway disturbances and their cell type-specific variations, especially in glia, is poorly understood. We integrated ALS GWAS-linked gene networks with human astrocyte-specific multi-omics datasets to elucidate pathognomonic signatures. It predicts that KIF5A, a motor protein kinesin-1 heavy-chain isoform, previously detected only in neurons, can also potentiate disease pathways in astrocytes. Using postmortem tissue and super-resolution structured illumination microscopy in cell-based perturbation platforms, we provide evidence that KIF5A is present in astrocyte processes and its deficiency disrupts structural integrity and mitochondrial transport. We show that this may underly cytoskeletal and trafficking changes in SOD1 ALS astrocytes characterised by low KIF5A levels, which can be rescued by c-Jun N-terminal Kinase-1 (JNK1), a kinesin transport regulator. Altogether, our pipeline reveals a mechanism controlling astrocyte process integrity, a pre-requisite for synapse maintenance and suggests a targetable loss-of-function in ALS.
Collapse
Affiliation(s)
- Kornélia Szebényi
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0PY, UK
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | | | - George M Gibbons
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0PY, UK
| | - Luca Biasetti
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Pedro Beltrao
- European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, CB10 1SD, UK.
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, 8093, Switzerland.
| | - András Lakatos
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0PY, UK.
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK.
| |
Collapse
|
26
|
Ho NJ, Chen X, Lei Y, Gu S. Decoding hereditary spastic paraplegia pathogenicity through transcriptomic profiling. Zool Res 2023; 44:650-662. [PMID: 37161652 PMCID: PMC10236304 DOI: 10.24272/j.issn.2095-8137.2022.281] [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: 12/30/2022] [Accepted: 05/10/2023] [Indexed: 05/11/2023] Open
Abstract
Hereditary spastic paraplegia (HSP) is a group of genetic motor neuron diseases resulting from length-dependent axonal degeneration of the corticospinal upper motor neurons. Due to the advancement of next-generation sequencing, more than 70 novel HSP disease-causing genes have been identified in the past decade. Despite this, our understanding of HSP physiopathology and the development of efficient management and treatment strategies remain poor. One major challenge in studying HSP pathogenicity is selective neuronal vulnerability, characterized by the manifestation of clinical symptoms that are restricted to specific neuronal populations, despite the presence of germline disease-causing variants in every cell of the patient. Furthermore, disease genes may exhibit ubiquitous expression patterns and involve a myriad of different pathways to cause motor neuron degeneration. In the current review, we explore the correlation between transcriptomic data and clinical manifestations, as well as the importance of interspecies models by comparing tissue-specific transcriptomic profiles of humans and mice, expression patterns of different genes in the brain during development, and single-cell transcriptomic data from related tissues. Furthermore, we discuss the potential of emerging single-cell RNA sequencing technologies to resolve unanswered questions related to HSP pathogenicity.
Collapse
Affiliation(s)
- Nicolas James Ho
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao Chen
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, 310058 China
| | - Yong Lei
- School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, China
- The Chinese University of Hong Kong (Shenzhen), Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Guangdong 518172, China. E-mail:
| | - Shen Gu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Kunming Institute of Zoology Chinese Academy of Sciences, The Chinese University of Hong Kong Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China. E-mail:
| |
Collapse
|
27
|
Ziff OJ, Neeves J, Mitchell J, Tyzack G, Martinez-Ruiz C, Luisier R, Chakrabarti AM, McGranahan N, Litchfield K, Boulton SJ, Al-Chalabi A, Kelly G, Humphrey J, Patani R. Integrated transcriptome landscape of ALS identifies genome instability linked to TDP-43 pathology. Nat Commun 2023; 14:2176. [PMID: 37080969 PMCID: PMC10119258 DOI: 10.1038/s41467-023-37630-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/22/2023] [Indexed: 04/22/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) causes motor neuron degeneration, with 97% of cases exhibiting TDP-43 proteinopathy. Elucidating pathomechanisms has been hampered by disease heterogeneity and difficulties accessing motor neurons. Human induced pluripotent stem cell-derived motor neurons (iPSMNs) offer a solution; however, studies have typically been limited to underpowered cohorts. Here, we present a comprehensive compendium of 429 iPSMNs from 15 datasets, and 271 post-mortem spinal cord samples. Using reproducible bioinformatic workflows, we identify robust upregulation of p53 signalling in ALS in both iPSMNs and post-mortem spinal cord. p53 activation is greatest with C9orf72 repeat expansions but is weakest with SOD1 and FUS mutations. TDP-43 depletion potentiates p53 activation in both post-mortem neuronal nuclei and cell culture, thereby functionally linking p53 activation with TDP-43 depletion. ALS iPSMNs and post-mortem tissue display enrichment of splicing alterations, somatic mutations, and gene fusions, possibly contributing to the DNA damage response.
Collapse
Affiliation(s)
- Oliver J Ziff
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, WC1N 3BG, UK.
| | - Jacob Neeves
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Jamie Mitchell
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Giulia Tyzack
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Carlos Martinez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Raphaelle Luisier
- Genomics and Health Informatics Group, Idiap Research Institute, Martigny, Switzerland
| | | | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Simon J Boulton
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Gavin Kelly
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Jack Humphrey
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rickie Patani
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, WC1N 3BG, UK.
| |
Collapse
|
28
|
Fatoki TH, Chukwuejim S, Udenigwe CC, Aluko RE. In Silico Exploration of Metabolically Active Peptides as Potential Therapeutic Agents against Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:5828. [PMID: 36982902 PMCID: PMC10058213 DOI: 10.3390/ijms24065828] [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: 01/31/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is regarded as a fatal neurodegenerative disease that is featured by progressive damage of the upper and lower motor neurons. To date, over 45 genes have been found to be connected with ALS pathology. The aim of this work was to computationally identify unique sets of protein hydrolysate peptides that could serve as therapeutic agents against ALS. Computational methods which include target prediction, protein-protein interaction, and peptide-protein molecular docking were used. The results showed that the network of critical ALS-associated genes consists of ATG16L2, SCFD1, VAC15, VEGFA, KEAP1, KIF5A, FIG4, TUBA4A, SIGMAR1, SETX, ANXA11, HNRNPL, NEK1, C9orf72, VCP, RPSA, ATP5B, and SOD1 together with predicted kinases such as AKT1, CDK4, DNAPK, MAPK14, and ERK2 in addition to transcription factors such as MYC, RELA, ZMIZ1, EGR1, TRIM28, and FOXA2. The identified molecular targets of the peptides that support multi-metabolic components in ALS pathogenesis include cyclooxygenase-2, angiotensin I-converting enzyme, dipeptidyl peptidase IV, X-linked inhibitor of apoptosis protein 3, and endothelin receptor ET-A. Overall, the results showed that AGL, APL, AVK, IIW, PVI, and VAY peptides are promising candidates for further study. Future work would be needed to validate the therapeutic properties of these hydrolysate peptides by in vitro and in vivo approaches.
Collapse
Affiliation(s)
- Toluwase Hezekiah Fatoki
- Department of Biochemistry, Federal University Oye-Ekiti, PMB 373, Oye 371104, Nigeria; (T.H.F.); (S.C.)
| | - Stanley Chukwuejim
- Department of Biochemistry, Federal University Oye-Ekiti, PMB 373, Oye 371104, Nigeria; (T.H.F.); (S.C.)
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Chibuike C. Udenigwe
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Rotimi E. Aluko
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Richardson Centre for Food Technology and Research, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
29
|
Udine E, Jain A, van Blitterswijk M. Advances in sequencing technologies for amyotrophic lateral sclerosis research. Mol Neurodegener 2023; 18:4. [PMID: 36635726 PMCID: PMC9838075 DOI: 10.1186/s13024-022-00593-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/23/2022] [Indexed: 01/14/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is caused by upper and lower motor neuron loss and has a fairly rapid disease progression, leading to fatality in an average of 2-5 years after symptom onset. Numerous genes have been implicated in this disease; however, many cases remain unexplained. Several technologies are being used to identify regions of interest and investigate candidate genes. Initial approaches to detect ALS genes include, among others, linkage analysis, Sanger sequencing, and genome-wide association studies. More recently, next-generation sequencing methods, such as whole-exome and whole-genome sequencing, have been introduced. While those methods have been particularly useful in discovering new ALS-linked genes, methodological advances are becoming increasingly important, especially given the complex genetics of ALS. Novel sequencing technologies, like long-read sequencing, are beginning to be used to uncover the contribution of repeat expansions and other types of structural variation, which may help explain missing heritability in ALS. In this review, we discuss how popular and/or upcoming methods are being used to discover ALS genes, highlighting emerging long-read sequencing platforms and their role in aiding our understanding of this challenging disease.
Collapse
Affiliation(s)
- Evan Udine
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224 USA ,grid.417467.70000 0004 0443 9942Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Road S, Jacksonville, FL 32224 USA
| | - Angita Jain
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224 USA ,grid.417467.70000 0004 0443 9942Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Road S, Jacksonville, FL 32224 USA ,grid.417467.70000 0004 0443 9942Center for Clinical and Translational Sciences, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224 USA
| | - Marka van Blitterswijk
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA.
| |
Collapse
|
30
|
Nam JY, Chun S, Lee TY, Seo Y, Kim K, Park J, Sung W, Oh KW, Lee S, Park JS, Oh J, Chung KC, An H, Chu HS, Son B, Kim SH. Long-term survival benefits of intrathecal autologous bone marrow-derived mesenchymal stem cells (Neuronata-R®: lenzumestrocel) treatment in ALS: Propensity-score-matched control, surveillance study. Front Aging Neurosci 2023; 15:1148444. [PMID: 37122380 PMCID: PMC10130504 DOI: 10.3389/fnagi.2023.1148444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
Objective Neuronata-R® (lenzumestrocel) is an autologous bone marrow-derived mesenchymal stem cell (BM-MSC) product, which was conditionally approved by the Korean Ministry of Food and Drug Safety (KMFDS, Republic of Korea) in 2013 for the treatment of amyotrophic lateral sclerosis (ALS). In the present study, we aimed to investigate the long-term survival benefits of treatment with intrathecal lenzumestrocel. Methods A total of 157 participants who received lenzumestrocel and whose symptom duration was less than 2 years were included in the analysis (BM-MSC group). The survival data of placebo participants from the Pooled-Resource Open-Access ALS Clinical Trials (PROACT) database were used as the external control, and propensity score matching (PSM) was used to reduce confounding biases in baseline characteristics. Adverse events were recorded during the entire follow-up period after the first treatment. Results Survival probability was significantly higher in the BM-MSC group compared to the external control group from the PROACT database (log-rank, p < 0.001). Multivariate Cox proportional hazard analysis showed a significantly lower hazard ratio for death in the BM-MSC group and indicated that multiple injections were more effective. Additionally, there were no serious adverse drug reactions found during the safety assessment, lasting a year after the first administration. Conclusion The results of the present study showed that lenzumestrocel treatment had a long-term survival benefit in real-world ALS patients.
Collapse
Affiliation(s)
- Jae-Yong Nam
- Central Research Center, CORESTEMCHEMON Inc., Seoul, Republic of Korea
| | - Sehwan Chun
- Central Research Center, CORESTEMCHEMON Inc., Seoul, Republic of Korea
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea
| | - Tae Yong Lee
- Central Research Center, CORESTEMCHEMON Inc., Seoul, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Yunjeong Seo
- Central Research Center, CORESTEMCHEMON Inc., Seoul, Republic of Korea
| | - Kwijoo Kim
- Central Research Center, CORESTEMCHEMON Inc., Seoul, Republic of Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Wonjae Sung
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Ki-Wook Oh
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Sanggon Lee
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Department of Neurology, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Republic of Korea
| | - Jin-Sung Park
- Department of Neurology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Juyeon Oh
- College of Nursing, Dankook University, Cheonan, Republic of Korea
| | - Kyung Cheon Chung
- Department of Neurology, Bethesda Gospel Hospital, Yangsan, Republic of Korea
| | - Hyonggin An
- Department of Biostatistics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyeon Sik Chu
- Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Bugyeong Son
- Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
- *Correspondence: Seung Hyun Kim,
| |
Collapse
|
31
|
Muzio L, Ghirelli A, Agosta F, Martino G. Novel therapeutic approaches for motor neuron disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:523-537. [PMID: 37620088 DOI: 10.1016/b978-0-323-98817-9.00027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that leads to the neurodegeneration and death of upper and lower motor neurons (MNs). Although MNs are the main cells involved in the process of neurodegeneration, a growing body of evidence points toward other cell types as concurrent to disease initiation and propagation. Given the current absence of effective therapies, the quest for other therapeutic targets remains open and still challenges the scientific community. Both neuronal and extra-neuronal mechanisms of cellular stress and damage have been studied and have posed the basis for the development of novel therapies that have been investigated on both animal models and humans. In this chapter, a thorough review of the main mechanisms of cellular damage and the respective therapeutic attempts targeting them is reported. The main areas covered include neuroinflammation, protein aggregation, RNA metabolism, and oxidative stress.
Collapse
Affiliation(s)
- Luca Muzio
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy
| | - Alma Ghirelli
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Agosta
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Gianvito Martino
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
32
|
Genetic Mapping of Behavioral Traits Using the Collaborative Cross Resource. Int J Mol Sci 2022; 24:ijms24010682. [PMID: 36614124 PMCID: PMC9821145 DOI: 10.3390/ijms24010682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
The complicated interactions between genetic background, environment and lifestyle factors make it difficult to study the genetic basis of complex phenotypes, such as cognition and anxiety levels, in humans. However, environmental and other factors can be tightly controlled in mouse studies. The Collaborative Cross (CC) is a mouse genetic reference population whose common genetic and phenotypic diversity is on par with that of humans. Therefore, we leveraged the power of the CC to assess 52 behavioral measures associated with locomotor activity, anxiety level, learning and memory. This is the first application of the CC in novel object recognition tests, Morris water maze tasks, and fear conditioning tests. We found substantial continuous behavioral variations across the CC strains tested, and mapped six quantitative trait loci (QTLs) which influenced these traits, defining candidate genetic variants underlying these QTLs. Overall, our findings highlight the potential of the CC population in behavioral genetic research, while the identified genomic loci and genes driving the variation of relevant behavioral traits provide a foundation for further studies.
Collapse
|
33
|
Zeng R, Wang J, Jiang R, Yang J, Zheng C, Wu H, Zhuo Z, Yang Q, Li J, Leung FW, Sha W, Chen H. Investigating Causality and Shared Genetic Architecture between Neurodegenerative Disorders and Inflammatory Bowel Disease. Aging Dis 2022:AD.2022.12209. [PMID: 37163440 PMCID: PMC10389839 DOI: 10.14336/ad.2022.12209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/09/2022] [Indexed: 05/12/2023] Open
Abstract
Published observational studies have revealed the connection between neurodegenerative disorders and inflammatory bowel disease (IBD), whereas the causal association remains largely unclear. Our study aims to assess the causality and identify the shared genetic architecture between neurodegenerative disorders and IBD. Two-sample Mendelian randomization analyses were performed to assess the causality between IBD and neurodegenerative disorders (amyotrophic lateral sclerosis [ALS], Alzheimer's disease [AD], Parkinson's disease [PD], and multiple sclerosis [MS]). Shared genetic loci, functional interpretation, and transcriptomic profiles were further investigated in ALS and IBD. We identified that genetic predisposition to IBD was suggestively associated with lower odds of ALS (odds ratio [OR] 0.96, 95% confidence interval [CI] 0.94 to 0.99). In contrast, IBD was not genetically associated with an increased risk of AD, PD, or MS (and vice versa). Two shared genetic loci (rs6571361 and rs7154847) were derived, and SCFD1, G2E3, and HEATR5A were further identified as novel risk genes with enriched functions related to membrane trafficking. G2E3 was differentially expressed and significantly correlated with SCFD1 in patients with ALS or IBD. Our study reveals the suggestively protective role of IBD on ALS, and does not support the causality of AD, PD, or MS on IBD (and vice versa). Our findings indicate possible shared genetic architecture and pathways between ALS and IBD. These results provide insights into the pathogenesis and therapeutics of IBD and neurodegenerative disorders.
Collapse
Affiliation(s)
- Ruijie Zeng
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jinghua Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Rui Jiang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jie Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Chunwen Zheng
- Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Huihuan Wu
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jingwei Li
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Felix W Leung
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Sepulveda Ambulatory Care Center, Veterans Affairs Greater Los Angeles Healthcare System, North Hills, California, USA
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
34
|
Analysis of SOD1 and C9orf72 mutations in patients with amyotrophic lateral sclerosis in Antioquia, Colombia. BIOMEDICA : REVISTA DEL INSTITUTO NACIONAL DE SALUD 2022; 42:623-632. [PMID: 36511680 DOI: 10.7705/biomedica.6060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis is a neurodegenerative disease with a possible multifactorial origin characterized by the progressive degeneration of motor neurons. There is a relatively high prevalence of this disease in Antioquia; however, there is no published genetic study to date in Colombia. Despite its unknown etiopathogenesis, more genetic risk factors possibly involved in the development of this disease are constantly found. OBJETIVES To evaluate G93A and D90A mutations in SOD1 gene and a short tandem repeat in C9orf72 within a cohort of amyotrophic lateral sclerosis patients from Antioquia, Colombia. Materials y methods: Thirty-four patients previously diagnosed with amyotrophic lateral sclerosis were included in the study. Peripheral blood samples were used for DNA extraction and genotyping. RESULTS No mutations were found in SOD1 (G93A and D90A) in any of the patients, while C9orf72 exhibited an allele with a statistically significant high prevalence in the study sample (8 hexanucleotide repeats of CAGCAG). CONCLUSIONS These results suggest an association between this short tandem repeat (STR) in C9orf72 and the presence of amyotrophic lateral sclerosis in the studied population. However, this association should be established in a larger sample size and with controls from the same population. In addition, there also seems to be a genetic anticipation effect for the disease regarding this locus, since patients with this genotype present an earlier onset.
Collapse
|
35
|
Wu Z, Sun J, Liao Z, Qiao J, Chen C, Ling C, Wang H. An update on the therapeutic implications of long-chain acyl-coenzyme A synthetases in nervous system diseases. Front Neurosci 2022; 16:1030512. [PMID: 36507355 PMCID: PMC9731139 DOI: 10.3389/fnins.2022.1030512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Long-chain acyl-coenzyme A synthetases (ACSLs) are a family of CoA synthetases that activate fatty acid (FA) with chain lengths of 12-20 carbon atoms by forming the acyl-AMP derivative in an isozyme-specific manner. This family mainly includes five members (ACSL1, ACSL3, ACSL4, ACSL5, and ACSL6), which are thought to have specific and different functions in FA metabolism and oxidative stress of mammals. Accumulating evidence shows that the dysfunction of ACSLs is likely to affect cell proliferation and lead to metabolic diseases in multiple organs and systems through different signaling pathways and molecular mechanisms. Hence, a central theme of this review is to emphasize the therapeutic implications of ACSLs in nervous system disorders.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi Liao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia Qiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China,*Correspondence: Hui Wang,
| |
Collapse
|
36
|
Vasilopoulou C, Duguez S, Duddy W. Genome-Wide Gene-Set Analysis Approaches in Amyotrophic Lateral Sclerosis. J Pers Med 2022; 12:1932. [PMID: 36422108 PMCID: PMC9699154 DOI: 10.3390/jpm12111932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 09/10/2024] Open
Abstract
The rapid increase in the number of genetic variants identified to be associated with Amyotrophic Lateral Sclerosis (ALS) through genome-wide association studies (GWAS) has created an emerging need to understand the functional pathways that are implicated in the pathology of ALS. Gene-set analysis (GSA) is a powerful method that can provide insight into the associated biological pathways, determining the joint effect of multiple genetic markers. The main contribution of this review is the collection of ALS GSA studies that employ GWAS or individual-based genotype data, investigating their methodology and results related to ALS-associated molecular pathways. Furthermore, the limitations in standard single-gene analyses are summarized, highlighting the power of gene-set analysis, and a brief overview of the statistical properties of gene-set analysis and related concepts is provided. The main aims of this review are to investigate the reproducibility of the collected studies and identify their strengths and limitations, in order to enhance the experimental design and therefore the quality of the results of future studies, deepening our understanding of this devastating disease.
Collapse
Affiliation(s)
| | | | - William Duddy
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT47 6SB, UK
| |
Collapse
|
37
|
Abstract
Macroautophagy/autophagy, a fundamental cell process for nutrient recycling and defense against pathogens (termed xenophagy), is crucial to human health. ATG16L2 (autophagy related 16 like 2) is an autophagic protein and a paralog of ATG16L1. Both proteins are implicated in similar diseases such as cancer and other chronic diseases; however, most autophagy studies to date have primarily focused on the function of ATG16L1, with ATG16L2 remaining uncharacterized and understudied. Overexpression of ATG16L2 has been reported in various cancers including colorectal, gastric, and prostate carcinomas, whereas altered methylation of ATG16L2 has been associated with lung cancer formation and poorer response to therapy in leukemia. In addition, ATG16L2 polymorphisms have been implicated in a range of other diseases including inflammatory bowel diseases and neurodegenerative disorders. Despite this likely role in human health, the function of this enigmatic protein in autophagy remains unknown. Here, we review current studies on ATG16L2 and collate evidence that suggests that this protein is a potential modulator of autophagy as well as the implications this has on pathogenesis.Abbreviations: ATG5: autophagy related 5; ATG12: autophagy related 12; ATG16L1: autophagy related 16 like 1; ATG16L2: autophagy related 16 like 2; CD: Crohn disease; IBD: inflammatory bowel diseases; IRGM: immunity related GTPase M; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PE: phosphatidylethanolamine; RB1CC1: RB1 inducible coiled-coil 1; SLE: systemic lupus erythematosus; WIPI2B: WD repeat domain, phosphoinositide interacting 2B.
Collapse
Affiliation(s)
- Laurence Don Wai Luu
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia,CONTACT Laurence Don Wai Luu School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Nadeem O. Kaakoush
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Natalia Castaño-Rodríguez
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia,Natalia Castaño-Rodríguez School of Biotechnology and Biomolecular Sciences, Faculty of Science, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
38
|
Feldman EL, Goutman SA, Petri S, Mazzini L, Savelieff MG, Shaw PJ, Sobue G. Amyotrophic lateral sclerosis. Lancet 2022; 400:1363-1380. [PMID: 36116464 PMCID: PMC10089700 DOI: 10.1016/s0140-6736(22)01272-7] [Citation(s) in RCA: 416] [Impact Index Per Article: 138.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/24/2022] [Accepted: 06/23/2022] [Indexed: 01/07/2023]
Abstract
Amyotrophic lateral sclerosis is a fatal CNS neurodegenerative disease. Despite intensive research, current management of amyotrophic lateral sclerosis remains suboptimal from diagnosis to prognosis. Recognition of the phenotypic heterogeneity of amyotrophic lateral sclerosis, global CNS dysfunction, genetic architecture, and development of novel diagnostic criteria is clarifying the spectrum of clinical presentation and facilitating diagnosis. Insights into the pathophysiology of amyotrophic lateral sclerosis, identification of disease biomarkers and modifiable risks, along with new predictive models, scales, and scoring systems, and a clinical trial pipeline of mechanism-based therapies, are changing the prognostic landscape. Although most recent advances have yet to translate into patient benefit, the idea of amyotrophic lateral sclerosis as a complex syndrome is already having tangible effects in the clinic. This Seminar will outline these insights and discuss the status of the management of amyotrophic lateral sclerosis for the general neurologist, along with future prospects that could improve care and outcomes for patients with amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Eva L Feldman
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Stephen A Goutman
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Letizia Mazzini
- ALS Centre, Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy; Department of Neurology, University of Piemonte Orientale, Novara, Italy
| | - Masha G Savelieff
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Gen Sobue
- Department of Neurology, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
39
|
Rodríguez-Quiroz R, Valdebenito-Maturana B. SoloTE for improved analysis of transposable elements in single-cell RNA-Seq data using locus-specific expression. Commun Biol 2022; 5:1063. [PMID: 36202992 PMCID: PMC9537157 DOI: 10.1038/s42003-022-04020-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/21/2022] [Indexed: 11/08/2022] Open
Abstract
Transposable Elements (TEs) contribute to the repetitive fraction in almost every eukaryotic genome known to date, and their transcriptional activation can influence the expression of neighboring genes in healthy and disease states. Single cell RNA-Seq (scRNA-Seq) is a technical advance that allows the study of gene expression on a cell-by-cell basis. Although a current computational approach is available for the single cell analysis of TE expression, it omits their genomic location. Here we show SoloTE, a pipeline that outperforms the previous approach in terms of computational resources and by allowing the inclusion of locus-specific TE activity in scRNA-Seq expression matrixes. We then apply SoloTE to several datasets to reveal the repertoire of TEs that become transcriptionally active in different cell groups, and based on their genomic location, we predict their potential impact on gene expression. As our tool takes as input the resulting files from standard scRNA-Seq processing pipelines, we expect it to be widely adopted in single cell studies to help researchers discover patterns of cellular diversity associated with TE expression.
Collapse
Affiliation(s)
- Rocío Rodríguez-Quiroz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW ALS genetics are highly dynamic and of great interest for the ALS research community. Each year, by using ever-growing datasets and cutting-edge methodology, an array of novel ALS-associated genes and downstream pathomechanisms are discovered. The increasing plenty and complexity of insights warrants regular summary by-reviews. RECENT FINDINGS Most recent disease gene discoveries constitute the candidate and risk genes SPTLC1 , KANK1 , CAV1 , HTT , and WDR7 , as well as seven novel risk loci. Cell type and functional enrichment analyses enlighten the genetic basis of selective motor neuron vulnerability in ALS demonstrating high expression of ALS-associated genes in cortical motor neurons and highlight the pathogenic significance of cell-autonomous processes. Major pathomechanistic insights have been gained regarding known ALS genes/proteins, specifically C9orf72 , TDP43, ANXA11 , and KIF5A . The first ASO-based gene-specific therapy trials in familial forms of ALS have yielded equivocal results stressing the re-evaluation of pathomechanisms linked to SOD1 and C9orf72 mutations. SUMMARY The genetic and molecular basis of ALS is increasingly examined on single-cell resolution. In the past 2 years, the understanding of the downstream mechanisms of several ALS genes and TDP-43 proteinopathy has been considerably extended. These insights will result in novel gene specific therapy approaches for sporadic ALS and genetic subtypes.
Collapse
|
41
|
Caprariello AV, Adams DJ. The landscape of targets and lead molecules for remyelination. Nat Chem Biol 2022; 18:925-933. [PMID: 35995862 PMCID: PMC9773298 DOI: 10.1038/s41589-022-01115-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/18/2022] [Indexed: 12/24/2022]
Abstract
Remyelination, or the restoration of myelin sheaths around axons in the central nervous system, is a multi-stage repair process that remains a major need for millions of patients with multiple sclerosis and other diseases of myelin. Even into adulthood, rodents and humans can generate new myelin-producing oligodendrocytes, leading to the therapeutic hypothesis that enhancing remyelination could lessen disease burden in multiple sclerosis. Multiple labs have used phenotypic screening to identify dozens of drugs that enhance oligodendrocyte formation, and several hit molecules have now advanced to clinical evaluation. Target identification studies have revealed that a large majority of these hits share the ability to inhibit a narrow range of cholesterol pathway enzymes and thereby induce cellular accumulation of specific sterol precursors to cholesterol. This Perspective surveys the recent fruitful intersection of chemical biology and remyelination and suggests multiple approaches toward new targets and lead molecules to promote remyelination.
Collapse
Affiliation(s)
| | - Drew J Adams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
42
|
Fatoki T, Chukwuejim S, Ibraheem O, Oke C, Ejimadu B, Olaoye I, Oyegbenro O, Salami T, Basorun R, Oluwadare O, Salawudeen Y. Harmine and 7,8-dihydroxyflavone synergistically suitable for amyotrophic lateral sclerosis management: An in silico study. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.83332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease characterized by progressive degeneration of both upper and lower motor neurons, resulting in paralysis and eventually leads to death from respiratory failure typically within 3 to 5 years of symptom onset. The aim of this work was to predict the pharmacokinetics and identify unique protein targets that are associated with potential anti-ALS phytochemicals and FDA-approved drugs, by in silico approaches.
Materials and methods: Standard computational tools (webserver and software) were used, and the methods used are clustering analysis, pharmacokinetics and molecular target predictions, and molecular docking simulation.
Results and discussion: The results show that riluzole, β-asarone, cryptotanshinone, harmine and 7,8-dihydroxyflavone have similar pharmacokinetics properties. Riluzole and harmine show 95% probability of target on norepinephrine transporter. Huperzine-A and cryptotanshinone show 100% probability of target on acetylcholinesterase. 7,8-dihydroxyflavone shows 35% probability of target on several carbonic anhydrases, 40% probability of target on CYP19A1, and 100% probability of target on inhibitor of nuclear factor kappa B kinase beta subunit and neurotrophic tyrosine kinase receptor type 2, respectively. Harmine also shows 95% probability of target on dual specificity tyrosine-phosphorylation-regulated kinases, threonine-protein kinases (haspin and PIM3), adrenergic receptors, cyclin-dependent kinases (CDK5 and CDK9), monoamine oxidase A, casein kinase I delta, serotonin receptors, dual specificity protein kinases (CLK1, CLK2, and CLK4), and nischarin, respectively. Also, the results of gene expression network show possible involvement of CDK1, CDK2, CDK4, ERK1, ERK2 and MAPK14 signaling pathways. This study shows that riluzole and harmine have closely similar physicochemical and pharmacokinetics properties as well as molecular targets, such as norepinephrine transporter (SLC6A2). Harmine, huperzine-A and cryptotanshinone could modulate acetylcholinesterase (AChE), which is involved in ALS-pathogenesis. The impact of 7,8-dihydroxyflavone on several carbonic anhydrases (CA) I, II, VII, IX, XII, and XIV, as well as CYP19A1, could help in remediating the respiratory failure associated with ALS.
Conclusion: Overall, harmine is found to be superior to riluzole, and the combination of harmine with 7,8-dihydroxyflavone can provide more effective treatment for ALS than the current regime. Further work is needed to validate the predicted therapeutic targets of harmine identified in this study on ALS model or clinical trials, using in silico, in vitro and in vivo techniques.
Graphical abstract:
Collapse
|
43
|
Langston RG, Beilina A, Reed X, Kaganovich A, Singleton AB, Blauwendraat C, Gibbs JR, Cookson MR. Association of a common genetic variant with Parkinson's disease is mediated by microglia. Sci Transl Med 2022; 14:eabp8869. [PMID: 35895835 PMCID: PMC9809150 DOI: 10.1126/scitranslmed.abp8869] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Studies of multiple neurodegenerative disorders have identified many genetic variants that are associated with risk of disease throughout a lifetime. For example, Parkinson's disease (PD) risk is attributed in part to both coding mutations in the leucine-rich repeat kinase 2 (LRRK2) gene and to a common noncoding variation in the 5' region of the LRRK2 locus, as identified by genome-wide association studies (GWAS). However, the mechanisms linking GWAS variants to pathogenicity are largely unknown. Here, we found that the influence of PD-associated noncoding variation on LRRK2 expression is specifically propagated through microglia and not by other cell types that express LRRK2 in the human brain. We find microglia-specific regulatory chromatin regions that modulate the LRRK2 expression in human frontal cortex and substantia nigra and confirm these results in a human-induced pluripotent stem cell-derived microglia model. We showed, using a large-scale clustered regularly interspaced short palindromic repeats interference (CRISPRi) screen, that a regulatory DNA element containing the single-nucleotide variant rs6581593 influences the LRRK2 expression in microglia. Our study demonstrates that cell type should be considered when evaluating the role of noncoding variation in disease pathogenesis and sheds light on the mechanism underlying the association of the 5' region of LRRK2 with PD risk.
Collapse
Affiliation(s)
- R. G. Langston
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - A. Beilina
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - X. Reed
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - A. Kaganovich
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - A. B. Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - C. Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - J. R. Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - M. R. Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
44
|
Pan S, Liu X, Liu T, Zhao Z, Dai Y, Wang YY, Jia P, Liu F. Causal Inference of Genetic Variants and Genes in Amyotrophic Lateral Sclerosis. Front Genet 2022; 13:917142. [PMID: 35812739 PMCID: PMC9257137 DOI: 10.3389/fgene.2022.917142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal progressive multisystem disorder with limited therapeutic options. Although genome-wide association studies (GWASs) have revealed multiple ALS susceptibility loci, the exact identities of causal variants, genes, cell types, tissues, and their functional roles in the development of ALS remain largely unknown. Here, we reported a comprehensive post-GWAS analysis of the recent large ALS GWAS (n = 80,610), including functional mapping and annotation (FUMA), transcriptome-wide association study (TWAS), colocalization (COLOC), and summary data-based Mendelian randomization analyses (SMR) in extensive multi-omics datasets. Gene property analysis highlighted inhibitory neuron 6, oligodendrocytes, and GABAergic neurons (Gad1/Gad2) as functional cell types of ALS and confirmed cerebellum and cerebellar hemisphere as functional tissues of ALS. Functional annotation detected the presence of multiple deleterious variants at three loci (9p21.2, 12q13.3, and 12q14.2) and highlighted a list of SNPs that are potentially functional. TWAS, COLOC, and SMR identified 43 genes at 24 loci, including 23 novel genes and 10 novel loci, showing significant evidence of causality. Integrating multiple lines of evidence, we further proposed that rs2453555 at 9p21.2 and rs229243 at 14q12 functionally contribute to the development of ALS by regulating the expression of C9orf72 in pituitary and SCFD1 in skeletal muscle, respectively. Together, these results advance our understanding of the biological etiology of ALS, feed into new therapies, and provide a guide for subsequent functional experiments.
Collapse
Affiliation(s)
- Siyu Pan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xinxuan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Tianzi Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yin-Ying Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Peilin Jia
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- *Correspondence: Fan Liu, ; Peilin Jia,
| | - Fan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Fan Liu, ; Peilin Jia,
| |
Collapse
|
45
|
Alvia M, Aytan N, Spencer KR, Foster ZW, Rauf NA, Guilderson L, Robey I, Averill JG, Walker SE, Alvarez VE, Huber BR, Mathais R, Cormier KA, Nicks R, Pothast M, Labadorf A, Agus F, Alosco ML, Mez J, Kowall NW, McKee AC, Brady CB, Stein TD. MicroRNA Alterations in Chronic Traumatic Encephalopathy and Amyotrophic Lateral Sclerosis. Front Neurosci 2022; 16:855096. [PMID: 35663558 PMCID: PMC9160996 DOI: 10.3389/fnins.2022.855096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Repetitive head impacts (RHI) and traumatic brain injuries are risk factors for the neurodegenerative diseases chronic traumatic encephalopathy (CTE) and amyotrophic lateral sclerosis (ALS). ALS and CTE are distinct disorders, yet in some instances, share pathology, affect similar brain regions, and occur together. The pathways involved and biomarkers for diagnosis of both diseases are largely unknown. MicroRNAs (miRNAs) involved in gene regulation may be altered in neurodegeneration and be useful as stable biomarkers. Thus, we set out to determine associations between miRNA levels and disease state within the prefrontal cortex in a group of brain donors with CTE, ALS, CTE + ALS and controls. Of 47 miRNAs previously implicated in neurological disease and tested here, 28 (60%) were significantly different between pathology groups. Of these, 21 (75%) were upregulated in both ALS and CTE, including miRNAs involved in inflammatory, apoptotic, and cell growth/differentiation pathways. The most significant change occurred in miR-10b, which was significantly increased in ALS, but not CTE or CTE + ALS. Overall, we found patterns of miRNA expression that are common and unique to CTE and ALS and that suggest shared and distinct mechanisms of pathogenesis.
Collapse
Affiliation(s)
- Marcela Alvia
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Nurgul Aytan
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | | | | | | | | | - Ian Robey
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - James G. Averill
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - Sean E. Walker
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - Victor E. Alvarez
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Bertrand R. Huber
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Rebecca Mathais
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Kerry A. Cormier
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Raymond Nicks
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Morgan Pothast
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Adam Labadorf
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Filisia Agus
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Michael L. Alosco
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Jesse Mez
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Neil W. Kowall
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Ann C. McKee
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Christopher B. Brady
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Thor D. Stein
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
46
|
Houghton OH, Mizielinska S, Gomez-Suaga P. The Interplay Between Autophagy and RNA Homeostasis: Implications for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Front Cell Dev Biol 2022; 10:838402. [PMID: 35573690 PMCID: PMC9096704 DOI: 10.3389/fcell.2022.838402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/14/2022] [Indexed: 01/18/2023] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia are neurodegenerative disorders that lie on a disease spectrum, sharing genetic causes and pathology, and both without effective therapeutics. Two pathways that have been shown to play major roles in disease pathogenesis are autophagy and RNA homeostasis. Intriguingly, there is an increasing body of evidence suggesting a critical interplay between these pathways. Autophagy is a multi-stage process for bulk and selective clearance of malfunctional cellular components, with many layers of regulation. Although the majority of autophagy research focuses on protein degradation, it can also mediate RNA catabolism. ALS/FTD-associated proteins are involved in many stages of autophagy and autophagy-mediated RNA degradation, particularly converging on the clearance of persistent pathological stress granules. In this review, we will summarise the progress in understanding the autophagy-RNA homeostasis interplay and how that knowledge contributes to our understanding of the pathobiology of ALS/FTD.
Collapse
Affiliation(s)
- O H Houghton
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom.,UK Dementia Research Institute at King's College London, London, United Kingdom
| | - S Mizielinska
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom.,UK Dementia Research Institute at King's College London, London, United Kingdom
| | - P Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom.,Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| |
Collapse
|
47
|
TDP-43 pathology: from noxious assembly to therapeutic removal. Prog Neurobiol 2022; 211:102229. [DOI: 10.1016/j.pneurobio.2022.102229] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/08/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
|
48
|
Lotti F, Przedborski S. Motoneuron Diseases. ADVANCES IN NEUROBIOLOGY 2022; 28:323-352. [PMID: 36066831 DOI: 10.1007/978-3-031-07167-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Motoneuron diseases (MNDs) represent a heterogeneous group of progressive paralytic disorders, mainly characterized by the loss of upper (corticospinal) motoneurons, lower (spinal) motoneurons or, often both. MNDs can occur from birth to adulthood and have a highly variable clinical presentation, even within gene-positive forms, suggesting the existence of environmental and genetic modifiers. A combination of cell autonomous and non-cell autonomous mechanisms contributes to motoneuron degeneration in MNDs, suggesting multifactorial pathogenic processes.
Collapse
Affiliation(s)
- Francesco Lotti
- Departments of Neurology, Pathology & Cell Biology, and Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Serge Przedborski
- Departments of Neurology, Pathology & Cell Biology, and Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
49
|
Kumar R, Haider S. Protein network analysis to prioritize key genes in amyotrophic lateral sclerosis. IBRO Neurosci Rep 2021; 12:25-44. [PMID: 34918006 PMCID: PMC8669318 DOI: 10.1016/j.ibneur.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 12/05/2021] [Indexed: 12/18/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal disease, progressive nature characterizes by loss of both upper and lower motor neuron functions. One of the major challenge is to understand the mechanism of ALS multifactorial nature. We aimed to explore some key genes related to ALS through bioinformatics methods for its therapeutic intervention. Here, we applied a systems biology approach involving experimentally validated 148 ALS-associated proteins and construct ALS protein-protein interaction network (ALS-PPIN). The network was further statistically analysed and identified bottleneck-hubs. The network is also subjected to identify modules which could have similar functions. The interaction between the modules and bottleneck-hubs provides the functional regulatory role of the ALS mechanism. The ALS-PPIN demonstrated a hierarchical scale-free nature. We identified 17 bottleneck-hubs, in which CDC5L, SNW1, TP53, SOD1, and VCP were the high degree nodes (hubs) in ALS-PPIN. CDC5L was found to control highly cluster modules and play a vital role in the stability of the overall network followed by SNW1, TP53, SOD1, and VCP. HSPA5 and HSPA8 acting as a common connector for CDC5L and TP53 bottleneck-hubs. The functional and disease association analysis showed ALS has a strong correlation with mRNA processing, protein deubiquitination, and neoplasms, nervous system, immune system disease classes. In the future, biochemical investigation of the observed bottleneck-hubs and their interacting partners could provide a further understanding of their role in the pathophysiology of ALS. Amyotrophic Lateral Sclerosis protein-protein interaction network (ALS-PPIN) followed a hierarchical scale-free nature. We identified 17 bottleneck-hubs in the ALS-PPIN. Among bottleneck-hubs we found CDC5L, SNW1, TP53, SOD1, and VCP were the high degree nodes (hubs) in the ALS-PPIN. CDC5L is the effective communicator with all five modules in the ALS-PPIN and followed by SNW1 and TP53. Modules are highly associated with various disease classes like neoplasms, nervous systems and others.
Collapse
Key Words
- ALS
- ALS, Amyotrophic Lateral Sclerosis
- ALS-PPIN
- ALS-PPIN, Amyotrophic Lateral Sclerosis Protein-Protein Interaction Network
- ALSoD, Amyotrophic Lateral Sclerosis online database
- BC, Betweenness centrality
- Bn-H, Bottleneck-hub
- Bottleneck-hubs
- CDC5L
- CDC5L, Cell division cycle5-likeprotein
- FUS, Fused in sarcoma
- MCODE, Molecular Complex Detection
- MND, Motor neuron disease
- SMA, Spinal muscular atrophy
- SMN, Survival of motor neuron
- SNW1
- SNW1, SNW domain-containing protein 1
- SOD1
- SOD1, Superoxide dismutase
- TP53
- TP53, Tumor protein p53
- VCP
- VCP, Valosin containing protein
Collapse
Affiliation(s)
- Rupesh Kumar
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Sec-62, Uttar Pradesh, India
| | - Shazia Haider
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Sec-62, Uttar Pradesh, India
| |
Collapse
|
50
|
van Rheenen W, van der Spek RAA, Bakker MK, van Vugt JJFA, Hop PJ, Zwamborn RAJ, de Klein N, Westra HJ, Bakker OB, Deelen P, Shireby G, Hannon E, Moisse M, Baird D, Restuadi R, Dolzhenko E, Dekker AM, Gawor K, Westeneng HJ, Tazelaar GHP, van Eijk KR, Kooyman M, Byrne RP, Doherty M, Heverin M, Al Khleifat A, Iacoangeli A, Shatunov A, Ticozzi N, Cooper-Knock J, Smith BN, Gromicho M, Chandran S, Pal S, Morrison KE, Shaw PJ, Hardy J, Orrell RW, Sendtner M, Meyer T, Başak N, van der Kooi AJ, Ratti A, Fogh I, Gellera C, Lauria G, Corti S, Cereda C, Sproviero D, D'Alfonso S, Sorarù G, Siciliano G, Filosto M, Padovani A, Chiò A, Calvo A, Moglia C, Brunetti M, Canosa A, Grassano M, Beghi E, Pupillo E, Logroscino G, Nefussy B, Osmanovic A, Nordin A, Lerner Y, Zabari M, Gotkine M, Baloh RH, Bell S, Vourc'h P, Corcia P, Couratier P, Millecamps S, Meininger V, Salachas F, Mora Pardina JS, Assialioui A, Rojas-García R, Dion PA, Ross JP, Ludolph AC, Weishaupt JH, Brenner D, Freischmidt A, Bensimon G, Brice A, Durr A, Payan CAM, Saker-Delye S, Wood NW, Topp S, Rademakers R, Tittmann L, Lieb W, Franke A, Ripke S, Braun A, Kraft J, Whiteman DC, Olsen CM, Uitterlinden AG, Hofman A, Rietschel M, Cichon S, Nöthen MM, Amouyel P, Traynor BJ, Singleton AB, Mitne Neto M, Cauchi RJ, Ophoff RA, Wiedau-Pazos M, Lomen-Hoerth C, van Deerlin VM, Grosskreutz J, Roediger A, Gaur N, Jörk A, Barthel T, Theele E, Ilse B, Stubendorff B, Witte OW, Steinbach R, Hübner CA, Graff C, Brylev L, Fominykh V, Demeshonok V, Ataulina A, Rogelj B, Koritnik B, Zidar J, Ravnik-Glavač M, Glavač D, Stević Z, Drory V, Povedano M, Blair IP, Kiernan MC, Benyamin B, Henderson RD, Furlong S, Mathers S, McCombe PA, Needham M, Ngo ST, Nicholson GA, Pamphlett R, Rowe DB, Steyn FJ, Williams KL, Mather KA, Sachdev PS, Henders AK, Wallace L, de Carvalho M, Pinto S, Petri S, Weber M, Rouleau GA, Silani V, Curtis CJ, Breen G, Glass JD, Brown RH, Landers JE, Shaw CE, Andersen PM, Groen EJN, van Es MA, Pasterkamp RJ, Fan D, Garton FC, McRae AF, Davey Smith G, Gaunt TR, Eberle MA, Mill J, McLaughlin RL, Hardiman O, Kenna KP, Wray NR, Tsai E, Runz H, Franke L, Al-Chalabi A, Van Damme P, van den Berg LH, Veldink JH. Common and rare variant association analyses in amyotrophic lateral sclerosis identify 15 risk loci with distinct genetic architectures and neuron-specific biology. Nat Genet 2021; 53:1636-1648. [PMID: 34873335 PMCID: PMC8648564 DOI: 10.1038/s41588-021-00973-1] [Citation(s) in RCA: 272] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 10/18/2021] [Indexed: 02/01/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with a lifetime risk of one in 350 people and an unmet need for disease-modifying therapies. We conducted a cross-ancestry genome-wide association study (GWAS) including 29,612 patients with ALS and 122,656 controls, which identified 15 risk loci. When combined with 8,953 individuals with whole-genome sequencing (6,538 patients, 2,415 controls) and a large cortex-derived expression quantitative trait locus (eQTL) dataset (MetaBrain), analyses revealed locus-specific genetic architectures in which we prioritized genes either through rare variants, short tandem repeats or regulatory effects. ALS-associated risk loci were shared with multiple traits within the neurodegenerative spectrum but with distinct enrichment patterns across brain regions and cell types. Of the environmental and lifestyle risk factors obtained from the literature, Mendelian randomization analyses indicated a causal role for high cholesterol levels. The combination of all ALS-associated signals reveals a role for perturbations in vesicle-mediated transport and autophagy and provides evidence for cell-autonomous disease initiation in glutamatergic neurons.
Collapse
Affiliation(s)
- Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Rick A A van der Spek
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Mark K Bakker
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joke J F A van Vugt
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Paul J Hop
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ramona A J Zwamborn
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Niek de Klein
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Harm-Jan Westra
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Olivier B Bakker
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Patrick Deelen
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gemma Shireby
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Eilis Hannon
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Matthieu Moisse
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Denis Baird
- Translational Biology, Biogen, Boston, MA, USA
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol, UK
| | - Restuadi Restuadi
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | | | - Annelot M Dekker
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Klara Gawor
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gijs H P Tazelaar
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Kristel R van Eijk
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Maarten Kooyman
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ross P Byrne
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Mark Doherty
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Mark Heverin
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Nicola Ticozzi
- Department of Neurology, Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
- Department of Pathophysiology and Transplantation, 'Dino Ferrari' Center, Università degli Studi di Milano, Milan, Italy
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Bradley N Smith
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marta Gromicho
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Siddharthan Chandran
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Suvankar Pal
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Karen E Morrison
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Richard W Orrell
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Thomas Meyer
- Charité University Hospital, Humboldt University, Berlin, Germany
| | - Nazli Başak
- Koç University, School of Medicine, KUTTAM-NDAL, Istanbul, Turkey
| | | | - Antonia Ratti
- Department of Neurology, Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Isabella Fogh
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan, Italy
| | - Giuseppe Lauria
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', MIlan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation, 'Dino Ferrari' Center, Università degli Studi di Milano, Milan, Italy
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Daisy Sproviero
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Massimiliano Filosto
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Adriano Chiò
- 'Rita Levi Montalcini' Department of Neuroscience, ALS Centre, University of Torino, Turin, Italy
- Neurologia 1, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Andrea Calvo
- 'Rita Levi Montalcini' Department of Neuroscience, ALS Centre, University of Torino, Turin, Italy
- Neurologia 1, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Cristina Moglia
- 'Rita Levi Montalcini' Department of Neuroscience, ALS Centre, University of Torino, Turin, Italy
- Neurologia 1, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Maura Brunetti
- 'Rita Levi Montalcini' Department of Neuroscience, ALS Centre, University of Torino, Turin, Italy
| | - Antonio Canosa
- 'Rita Levi Montalcini' Department of Neuroscience, ALS Centre, University of Torino, Turin, Italy
- Neurologia 1, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Maurizio Grassano
- 'Rita Levi Montalcini' Department of Neuroscience, ALS Centre, University of Torino, Turin, Italy
| | - Ettore Beghi
- Laboratory of Neurological Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elisabetta Pupillo
- Laboratory of Neurological Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giancarlo Logroscino
- Department of Clinical Research in Neurology, University of Bari at 'Pia Fondazione Card G. Panico' Hospital, Bari, Italy
| | - Beatrice Nefussy
- Neuromuscular Diseases Unit, Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Alma Osmanovic
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Essener Zentrum für Seltene Erkrankungen (EZSE), University Hospital Essen, Essen, Germany
| | - Angelica Nordin
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Yossef Lerner
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, the Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| | - Michal Zabari
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, the Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| | - Marc Gotkine
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, the Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| | - Robert H Baloh
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Neuromuscular Division, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shaughn Bell
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Neuromuscular Division, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Patrick Vourc'h
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, Tours, France
- UMR 1253, Université de Tours, Inserm, Tours, France
| | - Philippe Corcia
- UMR 1253, Université de Tours, Inserm, Tours, France
- Centre de référence sur la SLA, CHU de Tours, Tours, France
| | - Philippe Couratier
- Centre de référence sur la SLA, CHRU de Limoges, Limoges, France
- UMR 1094, Université de Limoges, Inserm, Limoges, France
| | - Stéphanie Millecamps
- ICM, Institut du Cerveau, Inserm, CNRS, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | | | - François Salachas
- ICM, Institut du Cerveau, Inserm, CNRS, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
- Département de Neurologie, Centre de référence SLA Ile de France, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | | | - Abdelilah Assialioui
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ricardo Rojas-García
- MND Clinic, Neurology Department, Hospital de la Santa Creu i Sant Pau de Barcelona, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Patrick A Dion
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Jay P Ross
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | | | - Jochen H Weishaupt
- Division of Neurodegeneration, Department of Neurology, University Medicine Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - David Brenner
- Division of Neurodegeneration, Department of Neurology, University Medicine Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Axel Freischmidt
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, Ulm, Germany
| | - Gilbert Bensimon
- Département de Pharmacologie Clinique, Hôpital de la Pitié-Salpêtrière, UPMC Pharmacologie, AP-HP, Paris, France
- Pharmacologie Sorbonne Université, Paris, France
- Institut du Cerveau, Paris Brain Institute ICM, Paris, France
- Laboratoire de Biostatistique, Epidémiologie Clinique, Santé Publique Innovation et Méthodologie (BESPIM), CHU-Nîmes, Nîmes, France
| | - Alexis Brice
- Sorbonne Université, Paris Brain Institute, APHP, INSERM, CNRS, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Alexandra Durr
- Sorbonne Université, Paris Brain Institute, APHP, INSERM, CNRS, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Christine A M Payan
- Département de Pharmacologie Clinique, Hôpital de la Pitié-Salpêtrière, UPMC Pharmacologie, AP-HP, Paris, France
| | | | - Nicholas W Wood
- Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Simon Topp
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Lukas Tittmann
- Popgen Biobank and Institute of Epidemiology, Christian Albrechts-University Kiel, Kiel, Germany
| | - Wolfgang Lieb
- Popgen Biobank and Institute of Epidemiology, Christian Albrechts-University Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
| | - Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
| | - David C Whiteman
- Cancer Control Group, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Catherine M Olsen
- Cancer Control Group, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Andre G Uitterlinden
- Department of Internal Medicine, Genetics Laboratory, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marcella Rietschel
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Central Institute of Mental Health, Mannheim, Germany
| | - Sven Cichon
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life and Brain Center, Bonn, Germany
- Division of Medical Genetics, University Hospital Basel and Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Neuroscience and Medicine INM-1, Research Center Juelich, Juelich, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life and Brain Center, Bonn, Germany
| | - Philippe Amouyel
- INSERM UMR1167-RID-AGE LabEx DISTALZ-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Centre Hospitalier of the University of Lille, Institut Pasteur de Lille, Lille, France
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Porter Neuroscience Research Center, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew B Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Porter Neuroscience Research Center, Bethesda, MD, USA
| | | | - Ruben J Cauchi
- Centre for Molecular Medicine and Biobanking and Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Roel A Ophoff
- University Medical Center Utrecht, Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, Utrecht, the Netherlands
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Martina Wiedau-Pazos
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | - Vivianna M van Deerlin
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Julian Grosskreutz
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
- Precision Neurology Unit, Department of Neurology, University Hospital Schleswig-Holstein, University of Luebeck, Luebeck, Germany
| | | | - Nayana Gaur
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Alexander Jörk
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Tabea Barthel
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Erik Theele
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Benjamin Ilse
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Robert Steinbach
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - Caroline Graff
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lev Brylev
- Department of Neurology, Bujanov Moscow Clinical Hospital, Moscow, Russia
- Moscow Research and Clinical Center for Neuropsychiatry of the Healthcare Department, Moscow, Russia
- Department of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology Russian Academy of Sciences, Moscow, Russia
| | - Vera Fominykh
- Department of Neurology, Bujanov Moscow Clinical Hospital, Moscow, Russia
- Department of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology Russian Academy of Sciences, Moscow, Russia
| | - Vera Demeshonok
- ALS-Care Center, 'GAOORDI', Medical Clinic of the St. Petersburg, St. Petersburg, Russia
| | - Anastasia Ataulina
- Department of Neurology, Bujanov Moscow Clinical Hospital, Moscow, Russia
| | - Boris Rogelj
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- Biomedical Research Institute BRIS, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Blaž Koritnik
- Ljubljana ALS Centre, Institute of Clinical Neurophysiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Janez Zidar
- Ljubljana ALS Centre, Institute of Clinical Neurophysiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Metka Ravnik-Glavač
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Damjan Glavač
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Zorica Stević
- Clinic of Neurology, Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vivian Drory
- Neuromuscular Diseases Unit, Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Monica Povedano
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ian P Blair
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Beben Benyamin
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Australian Centre for Precision Health and Allied Health and Human Performance, University of South Australia, Adelaide, South Australia, Australia
| | - Robert D Henderson
- Centre for Clinical Research, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Sarah Furlong
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Susan Mathers
- Calvary Health Care Bethlehem, Parkdale, Victoria, Australia
| | - Pamela A McCombe
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Merrilee Needham
- Fiona Stanley Hospital, Perth, Western Australia, Australia
- Notre Dame University, Fremantle, Western Australia, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Shyuan T Ngo
- Centre for Clinical Research, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Garth A Nicholson
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, New South Wales, Australia
- Molecular Medicine Laboratory, Concord Repatriation General Hospital, Concord, New South Wales, Australia
| | - Roger Pamphlett
- Discipline of Pathology and Department of Neuropathology, Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Dominic B Rowe
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Frederik J Steyn
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- The School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Kelly L Williams
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Neuroscience Research Australia Institute, Randwick, New South Wales, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, the Prince of Wales Hospital, UNSW, Randwick, New South Wales, Australia
| | - Anjali K Henders
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Mamede de Carvalho
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Pinto
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Markus Weber
- Neuromuscular Diseases Unit/ALS Clinic, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Guy A Rouleau
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Vincenzo Silani
- Department of Neurology, Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
- Department of Pathophysiology and Transplantation, 'Dino Ferrari' Center, Università degli Studi di Milano, Milan, Italy
| | - Charles J Curtis
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
- NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Gerome Breen
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
- NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Jonathan D Glass
- Department Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christopher E Shaw
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Peter M Andersen
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Ewout J N Groen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Michael A van Es
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Dongsheng Fan
- Department of Neurology, Third Hospital, Peking University, Beijing, China
| | - Fleur C Garton
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, Bristol, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, Bristol, UK
| | | | - Jonathan Mill
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Russell L McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kevin P Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Naomi R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Ellen Tsai
- Translational Biology, Biogen, Boston, MA, USA
| | - Heiko Runz
- Translational Biology, Biogen, Boston, MA, USA
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- King's College Hospital, London, UK
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|