74 results on '"Diane McKenna-Yasek"'
Search Results
2. The SOD1-mediated ALS phenotype shows a decoupling between age of symptom onset and disease duration
- Author
-
Sarah Opie-Martin, Alfredo Iacoangeli, Simon D. Topp, Olubunmi Abel, Keith Mayl, Puja R. Mehta, Aleksey Shatunov, Isabella Fogh, Harry Bowles, Naomi Limbachiya, Thomas P. Spargo, Ahmad Al-Khleifat, Kelly L. Williams, Jennifer Jockel-Balsarotti, Taha Bali, Wade Self, Lyndal Henden, Garth A. Nicholson, Nicola Ticozzi, Diane McKenna-Yasek, Lu Tang, Pamela J. Shaw, Adriano Chio, Albert Ludolph, Jochen H. Weishaupt, John E. Landers, Jonathan D. Glass, Jesus S. Mora, Wim Robberecht, Philip Van Damme, Russell McLaughlin, Orla Hardiman, Leonard van den Berg, Jan H. Veldink, Phillippe Corcia, Zorica Stevic, Nailah Siddique, Vincenzo Silani, Ian P. Blair, Dong-sheng Fan, Florence Esselin, Elisa de la Cruz, William Camu, Nazli A. Basak, Teepu Siddique, Timothy Miller, Robert H. Brown, Ammar Al-Chalabi, and Christopher E. Shaw
- Subjects
Science - Abstract
Abstract Superoxide dismutase (SOD1) gene variants may cause amyotrophic lateral sclerosis, some of which are associated with a distinct phenotype. Most studies assess limited variants or sample sizes. In this international, retrospective observational study, we compare phenotypic and demographic characteristics between people with SOD1-ALS and people with ALS and no recorded SOD1 variant. We investigate which variants are associated with age at symptom onset and time from onset to death or censoring using Cox proportional-hazards regression. The SOD1-ALS dataset reports age of onset for 1122 and disease duration for 883 people; the comparator population includes 10,214 and 9010 people respectively. Eight variants are associated with younger age of onset and distinct survival trajectories; a further eight associated with younger onset only and one with distinct survival only. Here we show that onset and survival are decoupled in SOD1-ALS. Future research should characterise rarer variants and molecular mechanisms causing the observed variability.
- Published
- 2022
- Full Text
- View/download PDF
3. Association of UBQLN1 mutation with Brown–Vialetto–Van Laere syndrome but not typical ALS
- Author
-
Paloma González-Pérez, Yubing Lu, Ru-Ju Chian, Peter C. Sapp, Rudolph E. Tanzi, Lars Bertram, Diane McKenna-Yasek, Fen-Biao Gao, and Robert H. Brown, Jr.
- Subjects
Amyotrophic lateral sclerosis ,Drosophila motor neuron disease ,TDP-43 ,Ubiquilins ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Genetic variants in UBQLN1 gene have been linked to neurodegeneration and mutations in UBQLN2 have recently been identified as a rare cause of amyotrophic lateral sclerosis (ALS). Objective: To test if genetic variants in UBQLN1 are involved in ALS. Methods: 102 and 94 unrelated patients with familial and sporadic forms of ALS were screened for UBQLN1 gene mutations. Single nucleotide variants were further screened in a larger set of sporadic ALS (SALS) patients and unrelated control subjects using high-throughput Taqman genotyping; variants were further assessed for novelty using the 1000Genomes and NHLBI databases. In vitro studies tested the effect of UBQLN1 variants on the ubiquitin–proteasome system (UPS). Results: Only two UBQLN1 coding variants were detected in the familial and sporadic ALS DNA set; one, the missense mutation p.E54D, was identified in a single patient with atypical motor neuron disease consistent with Brown–Vialetto–Van Laere syndrome (BVVLS), for whom c20orf54 mutations had been excluded. Functional studies revealed that UBQLN1E54D protein forms cytosolic aggregates that contain mislocalized TDP-43 and impairs degradation of ubiquitinated proteins through the proteasome. Conclusions: Genetic variants in UBQLN1 are not commonly associated with ALS. A novel UBQLN1 mutation (E45D) detected in a patient with BVVLS altered nuclear TDP-43 localization in vitro, suggesting that UPS dysfunction may also underlie the pathogenesis of this condition.
- Published
- 2012
- Full Text
- View/download PDF
4. AAV gene therapy for Tay-Sachs disease
- Author
-
Terence R. Flotte, Oguz Cataltepe, Ajit Puri, Ana Rita Batista, Richard Moser, Diane McKenna-Yasek, Catherine Douthwright, Gwladys Gernoux, Meghan Blackwood, Christian Mueller, Phillip W. L. Tai, Xuntian Jiang, Scot Bateman, Spiro G. Spanakis, Julia Parzych, Allison M. Keeler, Aly Abayazeed, Saurabh Rohatgi, Laura Gibson, Robert Finberg, Bruce A. Barton, Zeynep Vardar, Mohammed Salman Shazeeb, Matthew Gounis, Cynthia J. Tifft, Florian S. Eichler, Robert H. Brown, Douglas R. Martin, Heather L. Gray-Edwards, and Miguel Sena-Esteves
- Subjects
Tay-Sachs Disease ,Humans ,Anticonvulsants ,Genetic Therapy ,General Medicine ,Dependovirus ,General Biochemistry, Genetics and Molecular Biology - Abstract
Tay-Sachs disease (TSD) is an inherited neurological disorder caused by deficiency of hexosaminidase A (HexA). Here, we describe an adeno-associated virus (AAV) gene therapy expanded-access trial in two patients with infantile TSD (IND 18225) with safety as the primary endpoint and no secondary endpoints. Patient TSD-001 was treated at 30 months with an equimolar mix of AAVrh8-HEXA and AAVrh8-HEXB administered intrathecally (i.t.), with 75% of the total dose (1 × 10
- Published
- 2022
- Full Text
- View/download PDF
5. Suppression of mutant C9orf72 expression by a potent mixed backbone antisense oligonucleotide
- Author
-
Hélène Tran, Michael P. Moazami, Huiya Yang, Diane McKenna-Yasek, Catherine L. Douthwright, Courtney Pinto, Jake Metterville, Minwook Shin, Nitasha Sanil, Craig Dooley, Ajit Puri, Alexandra Weiss, Nicholas Wightman, Heather Gray-Edwards, Miklos Marosfoi, Robert M. King, Thomas Kenderdine, Daniele Fabris, Robert Bowser, Jonathan K. Watts, and Robert H. Brown
- Subjects
Neurons ,Mice ,C9orf72 Protein ,Mutation ,Animals ,Humans ,Mice, Transgenic ,General Medicine ,Fibroblasts ,Oligonucleotides, Antisense ,Article ,General Biochemistry, Genetics and Molecular Biology - Abstract
Expansions of a G(4)C(2) repeat in the C9ORF72 gene are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two devastating adult-onset neurodegenerative disorders. Using C9-ALS/FTD patient derived cells and C9ORF72 BAC transgenic mice, we have generated and optimized antisense oligonucleotides (ASOs) that selectively blunt expression of G(4)C(2) repeat containing transcripts and effectively suppress tissue levels of polyGP dipeptides. ASOs with reduced phosphorothioate content showed improved tolerability without sacrificing efficacy. In a single patient harboring mutant C9ORF72 with the G(4)C(2) repeat expansion, repeated dosing by intrathecal delivery of the optimal ASO was well tolerated, leading to significant reductions in levels of CSF polyGP. This report provides insight into the impact of nucleic acid chemistry on toxicity and for the first time demonstrates the feasibility of clinical suppression of the C9ORF72 gene. Further clinical trials will be required to demonstrate safety and efficacy of this therapy in patients with C9ORF72 gene mutations.
- Published
- 2021
- Full Text
- View/download PDF
6. SOD1Suppression with Adeno-Associated Virus and MicroRNA in Familial ALS
- Author
-
Nicholas Wightman, Merit Cudkowicz, Margaret A. Owegi, Dario Gelevski, Gwladys Gernoux, Meghan Blackwood, Christian Mueller, James D. Berry, Sarah Luppino, Catherine Douthwright, Robert H. Brown, Terrence R. Flotte, Derek H. Oakley, Diane McKenna-Yasek, Lindsay Pothier, and Matthew P. Frosch
- Subjects
medicine.medical_specialty ,business.industry ,SOD1 ,nutritional and metabolic diseases ,Autopsy ,General Medicine ,030204 cardiovascular system & hematology ,medicine.disease_cause ,medicine.disease ,Spinal cord ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,medicine.anatomical_structure ,Internal medicine ,medicine ,Gene silencing ,030212 general & internal medicine ,Amyotrophic lateral sclerosis ,Complication ,business ,Adeno-associated virus - Abstract
Two patients with familial amyotrophic lateral sclerosis (ALS) and mutations in the gene encoding superoxide dismutase 1 (SOD1) were treated with a single intrathecal infusion of adeno-associated virus encoding a microRNA targeting SOD1. In Patient 1, SOD1 levels in spinal cord tissue as analyzed on autopsy were lower than corresponding levels in untreated patients with SOD1-mediated ALS and in healthy controls. Levels of SOD1 in cerebrospinal fluid were transiently and only slightly lower in Patient 1 but were not affected in Patient 2. In Patient 1, meningoradiculitis developed after the infusion; Patient 2 was pretreated with immunosuppressive drugs and did not have this complication. Patient 1 had transient improvement in the strength of his right leg, a measure that had been relatively stable throughout his disease course, but there was no change in his vital capacity. Patient 2 had stable scores on a composite measure of ALS function and a stable vital capacity during a 12-month period. This study showed that intrathecal microRNA can be used as a potential treatment for SOD1-mediated ALS.
- Published
- 2020
- Full Text
- View/download PDF
7. Single breath counting is an effective screening tool for forced vital capacity in ALS
- Author
-
Colin Quinn, Corey T. McMillan, Namita Goyal, Kelly Almasy, Diane McKenna-Yasek, Margaret A. Owegi, Catherine Douthwright, James D. Berry, and Robert H. Brown
- Subjects
2019-20 coronavirus outbreak ,Vital capacity ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,business.industry ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Amyotrophic Lateral Sclerosis ,Vital Capacity ,COVID-19 ,Single breath ,medicine.disease ,Neurology ,medicine ,Humans ,Screening tool ,Neurology (clinical) ,Amyotrophic lateral sclerosis ,Intensive care medicine ,business ,Pandemics - Abstract
Objective: To measure the correlation between single breath counting (SBC) and forced vital capacity (liters, FVCL) in amyotrophic lateral sclerosis (ALS) patients and to define the utility of SBC ...
- Published
- 2021
8. Prospective natural history study of C9orf72 ALS clinical characteristics and biomarkers
- Author
-
Sonia Boodram, Amber Salter, Robert H. Baloh, Diane McKenna-Yasek, Matthew B. Harms, Thomas J. Esparza, Theodore Hyman, Robert H. Brown, Caroline Drain, Nicholas Wightman, Carlos Cruchaga, Alzheimer’s Disease Neuroimaging Initiative, Leonard H. van den Berg, Toby A. Ferguson, Jan H. Veldink, Alexander Sherman, Michael A. van Es, Hong Yu, Catherine Douthwright, Jennifer Jockel-Balsarotti, Merit Cudkowicz, Alexander McCampbell, Margaret A. Owegi, Timothy M. Miller, Nazem Atassi, Amber Malcolm, Alexander J. Cammack, Bálint S de Vries, and Jeffrey D. Rothstein
- Subjects
medicine.medical_specialty ,business.industry ,Clinical study design ,Retrospective cohort study ,DNA Repeat Expansion ,medicine.disease ,Natural history ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Medicine ,030212 general & internal medicine ,Neurology (clinical) ,Amyotrophic lateral sclerosis ,Age of onset ,business ,Prospective cohort study ,030217 neurology & neurosurgery ,Natural history study - Abstract
ObjectiveTo define the natural history of the C9orf72 amyotrophic lateral sclerosis (C9ALS) patient population, develop disease biomarkers, and characterize patient pathologies.MethodsWe prospectively collected clinical and demographic data from 116 symptomatic C9ALS and 12 non–amyotrophic lateral sclerosis (ALS) full expansion carriers across 7 institutions in the United States and the Netherlands. In addition, we collected blood samples for DNA repeat size assessment, CSF samples for biomarker identification, and autopsy samples for dipeptide repeat protein (DPR) size determination. Finally, we collected retrospective clinical data via chart review from 208 individuals with C9ALS and 450 individuals with singleton ALS.ResultsThe mean age at onset in the symptomatic prospective cohort was 57.9 ± 8.3 years, and median duration of survival after onset was 36.9 months. The monthly change was −1.8 ± 1.7 for ALS Functional Rating Scale–Revised and −1.4% ± 3.24% of predicted for slow vital capacity. In blood DNA, we found that G4C2 repeat size correlates positively with age. In CSF, we observed that concentrations of poly(GP) negatively correlate with DNA expansion size but do not correlate with measures of disease progression. Finally, we found that size of poly(GP) dipeptides in the brain can reach large sizes similar to that of their DNA repeat derivatives.ConclusionsWe present a thorough investigation of C9ALS natural history, providing the basis for C9ALS clinical trial design. We found that clinical features of this genetic subset are less variant than in singleton ALS. In addition, we identified important correlations of C9ALS patient pathologies with clinical and demographic data.
- Published
- 2019
- Full Text
- View/download PDF
9. First-in-human AAV Gene Therapy for Tay-Sachs Disease
- Author
-
Ana Rita Batista, Mohammed Salman Shazeeb, Richard P. Moser, Robert H. Brown, Florian Eichler, Spiro Spanakis, Miguel Sena-Esteves, Laura Gibson, Matthew J. Gounis, Christian Mueller, Xuntian Jiang, Oguz Cataltepe, Allison M. Keeler, Bruce A. Barton, Catherine Douthwright, Diane McKenna-Yasek, Scot Bateman, Saurabh Rohatgi, Heather L. Gray-Edwards, Z Vardar, Aly Abayazeed, Terence R. Flotte, Julia Parzych, Robert W. Finberg, Meghan Blackwood, Douglas R. Martin, Gwladys Gernoux, and Ajit S. Puri
- Subjects
endocrine system ,business.industry ,Genetic enhancement ,Tay-Sachs disease ,Medicine ,First in human ,business ,medicine.disease ,Virology - Abstract
Tay-Sachs Disease (TSD) is an inherited neurological disorder caused by deficiency of hexosaminidase A (HexA). Preclinical work demonstrated safety and efficacy of CNS gene therapy using AAVrh8-HEXA/HEXB. Here we describe an expanded access trial in two patients with infantile TSD (IND 18225). Case TSD-001 demonstrated neurodevelopmental regression by 8 months of age and severe seizures by 1 year was treated at 30 months. An equimolar mix of AAVrh8-HEXA and AAVrh8-HEXB (now AXO-AAV-GM2) was administered intrathecally (IT), with 75% of the dose (1x1014vg) delivered to the cisterna magna and 25% at the thoraco-lumbar junction. The second patient (TSD-002) was treated at 7 months of age with 4.2x1013 vg by a combination of bilateral thalamic (0.18 mL; 1.5x1012vg per thalamus), and IT infusion (3.9x1013vg). Both patients underwent immunosuppression with sirolimus, corticosteroids, and rituximab. Injection procedures were well tolerated and have shown no vector-related adverse events to date. CSF HexA activity nearly doubled from baseline and remained stable. In TSD-002 (now 16 months of age), MRI showed stabilization of disease by 3 months post-injection and appeared to temporarily deviate from the natural history of infantile TSD but declined again 6 months post-treatment. TSD-001 (now 4.5 years of age remains seizure-free on the same anti-convulsant therapy as pre-therapy, but TSD-002 developed seizures between 13 and 17 months posttreatment (by 2 years of age). Administration of AXO-AAV-GM2 by IT and thalamic injections was safe, HexA activity increased in CSF and ongoing myelination was apparent in the younger patient treated at an early symptomatic stage. This study provides early safety and proof-of-concept in humans for treatment of TSD patients by AAV gene therapy.
- Published
- 2021
- Full Text
- View/download PDF
10. Potent Mixed Backbone Antisense Oligonucleotide Safety Suppressed Expression of Mutant C9ORF72 Transcripts and Polypeptides: First in Human Pilot Study
- Author
-
Ajit S. Puri, Helene Tran, Thomas Kenderdine, Robert Bowser, Dan Fabris, Huiya Yang, Catherine Douthwright, Robert M Brown, Jonathan K. Watts, Robert M. King, Alexandra Weiss, Jake Metterville, Heather L. Gray-Edwards, M Marosfoi, Michael P. Moazami, Diane McKenna-Yasek, Courtney Pinto, Minwook Shin, Craig Dooley, Nitasha Sanil, and Nichols Wightman
- Subjects
Text mining ,C9orf72 ,business.industry ,Antisense oligonucleotides ,Mutant ,First in human ,Biology ,business ,Molecular biology - Abstract
Expansions of a G4C2 repeat in the C9ORF72 gene are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two devastating adult-onset neurodegenerative disorders. Proposed disease mechanisms include a gain of toxic functions of the G4C2 repeats, implying that selective reduction in levels of the repeat-containing transcripts would represent a treatment strategy for this disorder. In the present study, using C9-ALS/FTD patient derived cells and C9ORF72 BAC transgenic mice, we have generated and optimized antisense oligonucleotides (ASOs) that selectively blunt expression of G4C2 repeat containing transcripts in both the sense and anti-sense strands of C9ORF72 and effectively suppress tissue levels of polyGP dipeptides. In a single patient harboring mutant C9ORF72 with the G4C2 repeat expressions, repeated dosing by intrathecal delivery of the optimal ASO was well tolerated, leading to significant reductions in levels of CSF polyGP.
- Published
- 2021
- Full Text
- View/download PDF
11. Pathogenic huntingtin repeat expansions in patients with frontotemporal dementia and amyotrophic lateral sclerosis
- Author
-
Ramita Dewan, Ruth Chia, Jinhui Ding, Richard A. Hickman, Thor D. Stein, Yevgeniya Abramzon, Sarah Ahmed, Marya S. Sabir, Makayla K. Portley, Arianna Tucci, Kristina Ibáñez, F.N.U. Shankaracharya, Pamela Keagle, Giacomina Rossi, Paola Caroppo, Fabrizio Tagliavini, Maria L. Waldo, Per M. Johansson, Christer F. Nilsson, James B. Rowe, Luisa Benussi, Giuliano Binetti, Roberta Ghidoni, Edwin Jabbari, Coralie Viollet, Jonathan D. Glass, Andrew B. Singleton, Vincenzo Silani, Owen A. Ross, Mina Ryten, Ali Torkamani, Toshiko Tanaka, Luigi Ferrucci, Susan M. Resnick, Stuart Pickering-Brown, Christopher B. Brady, Neil Kowal, John A. Hardy, Vivianna Van Deerlin, Jean Paul Vonsattel, Matthew B. Harms, Huw R. Morris, Raffaele Ferrari, John E. Landers, Adriano Chiò, J. Raphael Gibbs, Clifton L. Dalgard, Sonja W. Scholz, Bryan J. Traynor, Adelani Adeleye, Camille Alba, Dagmar Bacikova, Daniel N. Hupalo, Elisa McGrath Martinez, Harvey B. Pollard, Gauthaman Sukumar, Anthony R. Soltis, Meila Tuck, Xijun Zhang, Matthew D. Wilkerson, Bradley N. Smith, Nicola Ticozzi, Claudia Fallini, Athina Soragia Gkazi, Simon D. Topp, Jason Kost, Emma L. Scotter, Kevin P. Kenna, Jack W. Miller, Cinzia Tiloca, Caroline Vance, Eric W. Danielson, Claire Troakes, Claudia Colombrita, Safa Al-Sarraj, Elizabeth A. Lewis, Andrew King, Daniela Calini, Viviana Pensato, Barbara Castellotti, Jacqueline de Belleroche, Frank Baas, Anneloor L.M.A. ten Asbroek, Peter C. Sapp, Diane McKenna-Yasek, Russell L. McLaughlin, Meraida Polak, Seneshaw Asress, Jesús Esteban-Pérez, José Luis Muñoz-Blanco, Zorica Stevic, Sandra D’Alfonso, Letizia Mazzini, Giacomo P. Comi, Roberto Del Bo, Mauro Ceroni, Stella Gagliardi, Giorgia Querin, Cinzia Bertolin, Wouter van Rheenen, Frank P. Diekstra, Rosa Rademakers, Marka van Blitterswijk, Kevin B. Boylan, Giuseppe Lauria, Stefano Duga, Stefania Corti, Cristina Cereda, Lucia Corrado, Gianni Sorarù, Kelly L. Williams, Garth A. Nicholson, Ian P. Blair, Claire Leblond-Manry, Guy A. Rouleau, Orla Hardiman, Karen E. Morrison, Jan H. Veldink, Leonard H. van den Berg, Ammar Al-Chalabi, Hardev Pall, Pamela J. Shaw, Martin R. Turner, Kevin Talbot, Franco Taroni, Alberto García-Redondo, Zheyang Wu, Cinzia Gellera, Antonia Ratti, Robert H. Brown, Christopher E. Shaw, John C. Ambrose, Prabhu Arumugam, Emma L. Baple, Marta Bleda, Freya Boardman-Pretty, Jeanne M. Boissiere, Christopher R. Boustred, H. Brittain, Mark J. Caulfield, Georgia C. Chan, Clare E.H. Craig, Louise C. Daugherty, Anna de Burca, Andrew Devereau, Greg Elgar, Rebecca E. Foulger, Tom Fowler, Pedro Furió-Tarí, Joanne M. Hackett, Dina Halai, Angela Hamblin, Shirley Henderson, James E. Holman, Tim J.P. Hubbard, Rob Jackson, Louise J. Jones, Dalia Kasperaviciute, Melis Kayikci, Lea Lahnstein, Kay Lawson, Sarah E.A. Leigh, Ivonne U.S. Leong, Javier F. Lopez, Fiona Maleady-Crowe, Joanne Mason, Ellen M. McDonagh, Loukas Moutsianas, Michael Mueller, Nirupa Murugaesu, Anna C. Need, Chris A. Odhams, Christine Patch, Daniel Perez-Gil, Dimitris Polychronopoulos, John Pullinger, Tahrima Rahim, Augusto Rendon, Pablo Riesgo-Ferreiro, Tim Rogers, Kevin Savage, Kushmita Sawant, Richard H. Scott, Afshan Siddiq, Alexander Sieghart, Damian Smedley, Katherine R. Smith, Alona Sosinsky, William Spooner, Helen E. Stevens, Alexander Stuckey, Razvan Sultana, Ellen R.A. Thomas, Simon R. Thompson, Carolyn Tregidgo, Emma Walsh, Sarah A. Watters, Matthew J. Welland, Eleanor Williams, Katarzyna Witkowska, Suzanne M. Wood, Magdalena Zarowiecki, Sampath Arepalli, Pavan Auluck, Robert H. Baloh, Robert Bowser, Alexis Brice, James Broach, William Camu, John Cooper-Knock, Philippe Corcia, Carsten Drepper, Vivian E. Drory, Travis L. Dunckley, Faraz Faghri, Jennifer Farren, Eva Feldman, Mary Kay Floeter, Pietro Fratta, Glenn Gerhard, Summer B. Gibson, Stephen A. Goutman, Terry D. Heiman-Patterson, Dena G. Hernandez, Ben Hoover, Lilja Jansson, Freya Kamel, Janine Kirby, Neil W. Kowall, Hannu Laaksovirta, Francesco Landi, Isabelle Le Ber, Serge Lumbroso, Daniel JL. MacGowan, Nicholas J. Maragakis, Gabriele Mora, Kevin Mouzat, Liisa Myllykangas, Mike A. Nalls, Richard W. Orrell, Lyle W. Ostrow, Roger Pamphlett, Erik Pioro, Stefan M. Pulst, John M. Ravits, Alan E. Renton, Wim Robberecht, Ian Robey, Ekaterina Rogaeva, Jeffrey D. Rothstein, Michael Sendtner, Katie C. Sidle, Zachary Simmons, David J. Stone, Pentti J. Tienari, John Q. Trojanowski, Juan C. Troncoso, Miko Valori, Philip Van Damme, Ludo Van Den Bosch, Lorne Zinman, Diego Albani, Barbara Borroni, Alessandro Padovani, Amalia Bruni, Jordi Clarimon, Oriol Dols-Icardo, Ignacio Illán-Gala, Alberto Lleó, Adrian Danek, Daniela Galimberti, Elio Scarpini, Maria Serpente, Caroline Graff, Huei-Hsin Chiang, Behzad Khoshnood, Linn Öijerstedt, Christopher M. Morris, Benedetta Nacmias, Sandro Sorbi, Jorgen E. Nielsen, Lynne E. Hjermind, Valeria Novelli, Annibale A. Puca, Pau Pastor, Ignacio Alvarez, Monica Diez-Fairen, Miquel Aguilar, Robert Perneczky, Janine Diehl-Schimd, Mina Rossi, Agustin Ruiz, Mercè Boada, Isabel Hernández, Sonia Moreno-Grau, Johannes C. Schlachetzki, Dag Aarsland, Marilyn S. Albert, Johannes Attems, Matthew J. Barrett, Thomas G. Beach, Lynn M. Bekris, David A. Bennett, Lilah M. Besser, Eileen H. Bigio, Sandra E. Black, Bradley F. Boeve, Ryan C. Bohannan, Francesca Brett, Maura Brunetti, Chad A. Caraway, Jose-Alberto Palma, Andrea Calvo, Antonio Canosa, Dennis Dickson, Charles Duyckaerts, Kelley Faber, Tanis Ferman, Margaret E. Flanagan, Gianluca Floris, Tatiana M. Foroud, Juan Fortea, Ziv Gan-Or, Steve Gentleman, Bernardino Ghetti, Jesse Raphael Gibbs, Alison Goate, David Goldstein, Isabel González-Aramburu, Neill R. Graff-Radford, Angela K. Hodges, Heng-Chen Hu, Daniel Hupalo, Jon Infante, Alex Iranzo, Scott M. Kaiser, Horacio Kaufmann, Julia Keith, Ronald C. Kim, Gregory Klein, Rejko Krüger, Walter Kukull, Amanda Kuzma, Carmen Lage, Suzanne Lesage, James B. Leverenz, Giancarlo Logroscino, Grisel Lopez, Seth Love, Qinwen Mao, Maria Jose Marti, Elisa Martinez-McGrath, Mario Masellis, Eliezer Masliah, Patrick May, Ian McKeith, Marek-Marsel Mesulam, Edwin S. Monuki, Kathy L. Newell, Lucy Norcliffe-Kaufmann, Laura Palmer, Matthew Perkins, Olga Pletnikova, Laura Molina-Porcel, Regina H. Reynolds, Eloy Rodríguez-Rodríguez, Jonathan D. Rohrer, Pascual Sanchez-Juan, Clemens R. Scherzer, Geidy E. Serrano, Vikram Shakkottai, Ellen Sidransky, Nahid Tayebi, Alan J. Thomas, Bension S. Tilley, Ronald L. Walton, Randy Woltjer, Zbigniew K. Wszolek, Georgia Xiromerisiou, Chiara Zecca, Hemali Phatnani, Justin Kwan, Dhruv Sareen, James R. Broach, Ximena Arcila-Londono, Edward B. Lee, Neil A. Shneider, Ernest Fraenkel, Noah Zaitlen, James D. Berry, Andrea Malaspina, Gregory A. Cox, Leslie M. Thompson, Steve Finkbeiner, Efthimios Dardiotis, Timothy M. Miller, Siddharthan Chandran, Suvankar Pal, Eran Hornstein, Daniel J. MacGowan, Terry Heiman-Patterson, Molly G. Hammell, Nikolaos.A. Patsopoulos, Oleg Butovsky, Joshua Dubnau, Avindra Nath, Matt Harms, Eleonora Aronica, Mary Poss, Jennifer Phillips-Cremins, John Crary, Nazem Atassi, Dale J. Lange, Darius J. Adams, Leonidas Stefanis, Marc Gotkine, Suma Babu, Towfique Raj, Sabrina Paganoni, Ophir Shalem, Colin Smith, Bin Zhang, Brent Harris, Iris Broce, Vivian Drory, John Ravits, Corey McMillan, Vilas Menon, Lani Wu, Steven Altschuler, Khaled Amar, Neil Archibald, Oliver Bandmann, Erica Capps, Alistair Church, Jan Coebergh, Alyssa Costantini, Peter Critchley, Boyd CP. Ghosh, Michele T.M. Hu, Christopher Kobylecki, P. Nigel Leigh, Carl Mann, Luke A. Massey, Uma Nath, Nicola Pavese, Dominic Paviour, Jagdish Sharma, Jenny Vaughan, HUS Neurocenter, Neurologian yksikkö, Department of Neurosciences, Clinicum, Pentti Tienari / Principal Investigator, Parkinson's UK, Human Genetics, ARD - Amsterdam Reproduction and Development, ANS - Complex Trait Genetics, Pathology, ANS - Cellular & Molecular Mechanisms, AII - Inflammatory diseases, Universidad de Cantabria, Rowe, James [0000-0001-7216-8679], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Huntington's Disease ,Pathology ,amyotrophic lateral sclerosis ,Huntingtin ,Neurology ,1702 Cognitive Sciences ,International ALS/FTD Genomics Consortium ,Neurodegenerative ,frontotemporal dementia ,3124 Neurology and psychiatry ,0302 clinical medicine ,Medicine ,2.1 Biological and endogenous factors ,Psychology ,Amyotrophic lateral sclerosis ,Aetiology ,Alzheimer's Disease Related Dementias (ADRD) ,NYGC ALS Consortium ,Huntingtin Protein ,DNA Repeat Expansion ,General Neuroscience ,Frontotemporal Dementia (FTD) ,International FTD Genetics Consortium ,whole-genome sequencing ,Frontotemporal Dementia ,Neurological ,Cognitive Sciences ,Lewy body dementia ,huntingtin ,repeat expansions ,Amyotrophic Lateral Sclerosis ,Humans ,Mutation ,Whole Genome Sequencing ,Frontotemporal dementia ,Huntington’s disease ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,FALS Sequencing Consortium ,Article ,03 medical and health sciences ,Atrophy ,Rare Diseases ,American Genome Center ,Clinical Research ,mental disorders ,Genetics ,Acquired Cognitive Impairment ,Dementia ,PROSPECT Consortium ,Neurology & Neurosurgery ,Lewy body ,business.industry ,International LBD Genomics Consortium ,Neurosciences ,3112 Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,nutritional and metabolic diseases ,medicine.disease ,Brain Disorders ,nervous system diseases ,030104 developmental biology ,Genomics England Research Consortium ,1701 Psychology ,ALS ,business ,1109 Neurosciences ,030217 neurology & neurosurgery - Abstract
Hannu Laaksovirta konsortion jäsenenä. The Genomics England Research Consortium, The International ALS/FTD Genomics Consortium (iAFGC), The International FTD Genetics Consortium (IFGC), The International LBD Genomics Consortium (iLBDGC), The NYGC ALS Consortium, The PROSPECT Consortium,17 James B. Rowe,17 Luisa Benussi,18 Giuliano Binetti,18,19 Roberta Ghidoni,18 Edwin Jabbari,20,21 Coralie Viollet,22 Jonathan D. Glass,23 Andrew B. Singleton,24 Vincenzo Silani,25,26 Owen A. Ross,27 Mina Ryten,8,28,29 Ali Torkamani,30 Toshiko Tanaka,31 Luigi Ferrucci,31 Susan M. Resnick,32 We examined the role of repeat expansions in the pathogenesis of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) by analyzing whole-genome sequence data from 2,442 FTD/ALS patients, 2,599 Lewy body dementia (LBD) patients, and 3,158 neurologically healthy subjects. Pathogenic expansions (range, 40?64 CAG repeats) in the huntingtin (HTT) gene were found in three (0.12%) patients diagnosed with pure FTD/ALS syndromes but were not present in the LBD or healthy cohorts. We replicated our findings in an independent collection of 3,674 FTD/ALS patients. Postmortem evaluations of two patients revealed the classical TDP-43 pathology of FTD/ALS, as well as huntingtin-positive, ubiquitin-positive aggregates in the frontal cortex. The neostriatal atrophy that pathologically defines Huntington?s disease was absent in both cases. Our findings reveal an etiological relationship between HTT repeat expansions and FTD/ALS syndromes and indicate that genetic screening of FTD/ALS patients for HTT repeat expansions should be considered. We examined the role of repeat expansions in the pathogenesis of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) by analyzing whole-genome sequence data from 2,442 FTD/ALS patients, 2,599 Lewy body dementia (LBD) patients, and 3,158 neurologically healthy subjects. Pathogenic expansions (range, 40?64 CAG repeats) in the huntingtin (HTT) gene were found in three (0.12%) patients diagnosed with pure FTD/ALS syndromes but were not present in the LBD or healthy cohorts. We replicated our findings in an independent collection of 3,674 FTD/ALS patients. Postmortem evaluations of two patients revealed the classical TDP-43 pathology of FTD/ALS, as well as huntingtin-positive, ubiquitin-positive aggregates in the frontal cortex. The neostriatal atrophy that pathologically defines Huntington?s disease was absent in both cases. Our findings reveal an etiological relationship between HTT repeat expansions and FTD/ALS syndromes and indicate that genetic screening of FTD/ALS patients for HTT repeat expansions should be considered.
- Published
- 2020
- Full Text
- View/download PDF
12. Randomized trial of l-serine in patients with hereditary sensory and autonomic neuropathy type 1
- Author
-
Razina Aziz-Bose, Anne Louise Oaklander, Diane McKenna-Yasek, William S. David, Florian Eichler, Vera Fridman, Eric A. Macklin, Peter Novak, Thorsten Hornemann, Kailey Walsh, Robert H. Brown, Saranya Suriyanarayanan, University of Zurich, and Eichler, Florian
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Adolescent ,Neural Conduction ,Serine C-Palmitoyltransferase ,610 Medicine & health ,L serine ,Placebo ,Placebo group ,Article ,law.invention ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Double-Blind Method ,Randomized controlled trial ,law ,Surveys and Questionnaires ,Internal medicine ,540 Chemistry ,Hereditary sensory and autonomic neuropathy ,Serine ,medicine ,Humans ,In patient ,Hereditary Sensory and Autonomic Neuropathies ,Adverse effect ,10038 Institute of Clinical Chemistry ,Aged ,Pain Measurement ,Sphingolipids ,business.industry ,Middle Aged ,medicine.disease ,3. Good health ,2728 Neurology (clinical) ,Treatment Outcome ,030104 developmental biology ,Female ,Neurology (clinical) ,Autonomic neuropathy ,business ,Ubiquitin Thiolesterase ,030217 neurology & neurosurgery ,Follow-Up Studies - Abstract
ObjectiveTo evaluate the safety and efficacy of l-serine in humans with hereditary sensory autonomic neuropathy type I (HSAN1).MethodsIn this randomized, placebo-controlled, parallel-group trial with open-label extension, patients aged 18–70 years with symptomatic HSAN1 were randomized to l-serine (400 mg/kg/day) or placebo for 1 year. All participants received l-serine during the second year. The primary outcome measure was the Charcot-Marie-Tooth Neuropathy Score version 2 (CMTNS). Secondary outcomes included plasma sphingolipid levels, epidermal nerve fiber density, electrophysiologic measurements, patient-reported measures, and adverse events.ResultsBetween August 2013 and April 2014, we enrolled and randomized 18 participants, 16 of whom completed the study. After 1 year, the l-serine group experienced improvement in CMTNS relative to the placebo group (−1.5 units, 95% CI −2.8 to −0.1, p = 0.03), with evidence of continued improvement in the second year of treatment (−0.77, 95% CI −1.67 to 0.13, p = 0.09). Concomitantly, deoxysphinganine levels dropped in l-serine-treated but not placebo-treated participants (59% decrease vs 11% increase; p < 0.001). There were no serious adverse effects related to l-serine.ConclusionHigh-dose oral l-serine supplementation appears safe in patients with HSAN1 and is potentially effective at slowing disease progression.Clinicaltrials.gov identifierNCT01733407.Classification of evidenceThis study provides Class I evidence that high-dose oral l-serine supplementation significantly slows disease progression in patients with HSAN1.
- Published
- 2019
- Full Text
- View/download PDF
13. A Safe and Reliable Technique for CNS Delivery of AAV Vectors in the Cisterna Magna
- Author
-
Sundeep Chandra, Neil Aronin, Ajit S. Puri, Miguel Sena-Esteves, Matthew J. Gounis, Paul D. Gamlin, Heather L. Gray-Edwards, Chris Christou, Elise B. Diffie, Toloo Taghian, Stephanie G Bertrand, Miklos G. Marosfoi, Diane McKenna-Yasek, Deborah Fernau, Ana Rita Batista, Phillip W. L. Tai, Robert M. King, Douglas R. Martin, Tim Kuchel, Oguz Cataltepe, Terence R. Flotte, Anne S Maguire, and Raj Perumal
- Subjects
Central Nervous System ,Catheters ,Genetic enhancement ,Genetic Vectors ,Video Recording ,Gene Expression ,Gene delivery ,Bioinformatics ,Cisterna magna ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Genes, Reporter ,Transduction, Genetic ,Drug Discovery ,Cisterna Magna ,Genetics ,Lysosomal storage disease ,medicine ,Animals ,Humans ,In patient ,Transgenes ,Adverse effect ,Molecular Biology ,Injections, Spinal ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Sheep ,business.industry ,Gene Transfer Techniques ,Genetic Therapy ,Dependovirus ,medicine.disease ,Spinal cord ,Magnetic Resonance Imaging ,medicine.anatomical_structure ,Surgery, Computer-Assisted ,030220 oncology & carcinogenesis ,Models, Animal ,Molecular Medicine ,Original Article ,business ,Tomography, X-Ray Computed - Abstract
Global gene delivery to the CNS has therapeutic importance for the treatment of neurological disorders that affect the entire CNS. Due to direct contact with the CNS, cerebrospinal fluid (CSF) is an attractive route for CNS gene delivery. A safe and effective route to achieve global gene distribution in the CNS is needed, and administration of genes through the cisterna magna (CM) via a suboccipital puncture results in broad distribution in the brain and spinal cord. However, translation of this technique to clinical practice is challenging due to the risk of serious and potentially fatal complications in patients. Herein, we report development of a gene therapy delivery method to the CM through adaptation of an intravascular microcatheter, which can be safely navigated intrathecally under fluoroscopic guidance. We examined the safety, reproducibility, and distribution/transduction of this method in sheep using a self-complementary adeno-associated virus 9 (scAAV9)-GFP vector. This technique was used to treat two Tay-Sachs disease patients (30 months old and 7 months old) with AAV gene therapy. No adverse effects were observed during infusion or post-treatment. This delivery technique is a safe and minimally invasive alternative to direct infusion into the CM, achieving broad distribution of AAV gene transfer to the CNS.
- Published
- 2019
14. Correction of pseudoexon splicing caused by a novel intronic dysferlin mutation
- Author
-
Teresinha Evangelista, Diane McKenna-Yasek, Janice A. Dominov, Babi Ramesh Reddy Nallamilli, Judith N Hudson, Hanns Lochmüller, Virginie Kergourlay, Marc Bartoli, Madhuri Hegde, Özgün Uyan, Robert H. Brown, Nicolas Lévy, Laura E. Rufibach, Martin Krahn, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Département de génétique médicale [Hôpital de la Timone - APHM], Institut National de la Santé et de la Recherche Médicale (INSERM)- Hôpital de la Timone [CHU - APHM] (TIMONE)-Assistance Publique - Hôpitaux de Marseille (APHM)-Aix Marseille Université (AMU), Department of Neurology, Institute of Human Genetics, and Newcastle University [Newcastle]
- Subjects
0301 basic medicine ,Dysferlinopathy ,RNA Splicing ,Dysferlin ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Medicine ,Humans ,Research Articles ,ComputingMilieux_MISCELLANEOUS ,Genetics ,biology ,business.industry ,General Neuroscience ,Point mutation ,Intron ,Membrane Proteins ,Oligonucleotides, Antisense ,medicine.disease ,Exon skipping ,Introns ,3. Good health ,Distal Myopathies ,Muscular Atrophy ,030104 developmental biology ,Muscular Dystrophies, Limb-Girdle ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,RNA splicing ,Mutation ,biology.protein ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Limb-girdle muscular dystrophy ,Research Article - Abstract
Objective Dysferlin is a large transmembrane protein that functions in critical processes of membrane repair and vesicle fusion. Dysferlin‐deficiency due to mutations in the dysferlin gene leads to muscular dystrophy (Miyoshi myopathy (MM), limb girdle muscular dystrophy type 2B (LGMD2B), distal myopathy with anterior tibial onset (DMAT)), typically with early adult onset. At least 416 pathogenic dysferlin mutations are known, but for approximately 17% of patients, one or both of their pathogenic variants remain undefined following standard exon sequencing methods that interrogate exons and nearby flanking intronic regions but not the majority of intronic regions. Methods We sequenced RNA from myogenic cells to identify a novel dysferlin pathogenic variant in two affected siblings that previously had only one disease‐causing variant identified. We designed antisense oligonucleotides (AONs) to bypass the effects of this mutation on RNA splicing. Results We identified a new pathogenic point mutation deep within dysferlin intron 50i. This intronic variant causes aberrant mRNA splicing and inclusion of an additional pseudoexon (PE, we term PE50.1) within the mature dysferlin mRNA. PE50.1 inclusion alters the protein sequence, causing premature translation termination. We identified this mutation in 23 dysferlinopathy patients (seventeen families), revealing it to be one of the more prevalent dysferlin mutations. We used AON‐mediated exon skipping to correct the aberrant PE50.1 splicing events in vitro, which increased normal mRNA production and significantly restored dysferlin protein expression. Interpretation Deep intronic mutations can be a common underlying cause of dysferlinopathy, and importantly, could be treatable with AON‐based exon‐skipping strategies.
- Published
- 2019
- Full Text
- View/download PDF
15. NurOwn, phase 2, randomized, clinical trial in patients with ALS: Safety, clinical, and biomarker results
- Author
-
Anthony J. Windebank, Margaret A. Owegi, Nathan P. Staff, Diane McKenna-Yasek, Merit Cudkowicz, Yael Gothelf, James D. Berry, Haggai Kaspi, Katherine Nicholson, Robert H. Brown, Munish Mehra, Y.S. Levy, Natalie Abramov, and Revital Aricha
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Phases of clinical research ,Placebo ,Mesenchymal Stem Cell Transplantation ,Transplantation, Autologous ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,Double-Blind Method ,law ,Internal medicine ,Medicine ,Humans ,Nerve Growth Factors ,Amyotrophic lateral sclerosis ,Aged ,business.industry ,Amyotrophic Lateral Sclerosis ,Middle Aged ,medicine.disease ,3. Good health ,Transplantation ,Clinical trial ,030104 developmental biology ,Biomarker (medicine) ,Population study ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
ObjectiveTo determine the safety and efficacy of mesenchymal stem cell (MSC)-neurotrophic factor (NTF) cells (NurOwn®, autologous bone marrow-derived MSCs, induced to secrete NTFs) delivered by combined intrathecal and intramuscular administration to participants with amyotrophic lateral sclerosis (ALS) in a phase 2 randomized controlled trial.MethodsThe study enrolled 48 participants randomized 3:1 (treatment: placebo). After a 3-month pretransplant period, participants received 1 dose of MSC-NTF cells (n = 36) or placebo (n = 12) and were followed for 6 months. CSF was collected before and 2 weeks after transplantation.ResultsThe study met its primary safety endpoint. The rate of disease progression (Revised ALS Functional Rating Scale [ALSFRS-R] slope change) in the overall study population was similar in treated and placebo participants. In a prespecified rapid progressor subgroup (n = 21), rate of disease progression was improved at early time points (p < 0.05). To address heterogeneity, a responder analysis showed that a higher proportion of treated participants experienced ≥1.5 points/month ALSFRS-R slope improvement compared to placebo at all time points, and was significant in rapid progressors at 4 and 12 weeks (p = 0.004 and 0.046, respectively). CSF neurotrophic factors increased and CSF inflammatory biomarkers decreased in treated participants (p < 0.05) post-transplantation. CSF monocyte chemoattractant protein-1 levels correlated with ALSFRS-R slope improvement up to 24 weeks (p < 0.05).ConclusionA single-dose transplantation of MSC-NTF cells is safe and demonstrated early promising signs of efficacy. This establishes a clear path forward for a multidose randomized clinical trial of intrathecal autologous MSC-NTF cell transplantation in ALS.Classification of evidenceThis phase II study provides Class I evidence.
- Published
- 2019
16. Prospective natural history study of
- Author
-
Alexander J, Cammack, Nazem, Atassi, Theodore, Hyman, Leonard H, van den Berg, Matthew, Harms, Robert H, Baloh, Robert H, Brown, Michael A, van Es, Jan H, Veldink, Balint S, de Vries, Jeffrey D, Rothstein, Caroline, Drain, Jennifer, Jockel-Balsarotti, Amber, Malcolm, Sonia, Boodram, Amber, Salter, Nicholas, Wightman, Hong, Yu, Alexander V, Sherman, Thomas J, Esparza, Diane, McKenna-Yasek, Margaret A, Owegi, Catherine, Douthwright, Alexander, McCampbell, Toby, Ferguson, Carlos, Cruchaga, Merit, Cudkowicz, and Timothy M, Miller
- Subjects
Male ,Heterozygote ,DNA Repeat Expansion ,C9orf72 Protein ,Amyotrophic Lateral Sclerosis ,Middle Aged ,Article ,Humans ,Female ,Longitudinal Studies ,Prospective Studies ,Age of Onset ,Biomarkers ,Follow-Up Studies ,Retrospective Studies - Abstract
OBJECTIVE: To define the natural history of the C9orf72 amyotrophic lateral sclerosis (C9ALS) patient population, develop disease biomarkers, and characterize patient pathologies. METHODS: We prospectively collected clinical and demographic data from 116 symptomatic C9ALS and 12 non–amyotrophic lateral sclerosis (ALS) full expansion carriers across 7 institutions in the United States and the Netherlands. In addition, we collected blood samples for DNA repeat size assessment, CSF samples for biomarker identification, and autopsy samples for dipeptide repeat protein (DPR) size determination. Finally, we collected retrospective clinical data via chart review from 208 individuals with C9ALS and 450 individuals with singleton ALS. RESULTS: The mean age at onset in the symptomatic prospective cohort was 57.9 ± 8.3 years, and median duration of survival after onset was 36.9 months. The monthly change was −1.8 ± 1.7 for ALS Functional Rating Scale–Revised and −1.4% ± 3.24% of predicted for slow vital capacity. In blood DNA, we found that G(4)C(2) repeat size correlates positively with age. In CSF, we observed that concentrations of poly(GP) negatively correlate with DNA expansion size but do not correlate with measures of disease progression. Finally, we found that size of poly(GP) dipeptides in the brain can reach large sizes similar to that of their DNA repeat derivatives. CONCLUSIONS: We present a thorough investigation of C9ALS natural history, providing the basis for C9ALS clinical trial design. We found that clinical features of this genetic subset are less variant than in singleton ALS. In addition, we identified important correlations of C9ALS patient pathologies with clinical and demographic data.
- Published
- 2018
17. Genome-wide Analyses Identify KIF5A as a Novel ALS Gene
- Author
-
Aude Nicolas, Kevin P. Kenna, Alan E. Renton, Nicola Ticozzi, Faraz Faghri, Ruth Chia, Janice A. Dominov, Brendan J. Kenna, Mike A. Nalls, Pamela Keagle, Alberto M. Rivera, Wouter van Rheenen, Natalie A. Murphy, Joke J.F.A. van Vugt, Joshua T. Geiger, Rick A. Van der Spek, Hannah A. Pliner, null Shankaracharya, Bradley N. Smith, Giuseppe Marangi, Simon D. Topp, Yevgeniya Abramzon, Athina Soragia Gkazi, John D. Eicher, Aoife Kenna, Gabriele Mora, Andrea Calvo, Letizia Mazzini, Nilo Riva, Jessica Mandrioli, Claudia Caponnetto, Stefania Battistini, Paolo Volanti, Vincenzo La Bella, Francesca L. Conforti, Giuseppe Borghero, Sonia Messina, Isabella L. Simone, Francesca Trojsi, Fabrizio Salvi, Francesco O. Logullo, Sandra D’Alfonso, Lucia Corrado, Margherita Capasso, Luigi Ferrucci, Cristiane de Araujo Martins Moreno, Sitharthan Kamalakaran, David B. Goldstein, Aaron D. Gitler, Tim Harris, Richard M. Myers, Hemali Phatnani, Rajeeva Lochan Musunuri, Uday Shankar Evani, Avinash Abhyankar, Michael C. Zody, Julia Kaye, Steven Finkbeiner, Stacia K. Wyman, Alex LeNail, Leandro Lima, Ernest Fraenkel, Clive N. Svendsen, Leslie M. Thompson, Jennifer E. Van Eyk, James D. Berry, Timothy M. Miller, Stephen J. Kolb, Merit Cudkowicz, Emily Baxi, Michael Benatar, J. Paul Taylor, Evadnie Rampersaud, Gang Wu, Joanne Wuu, Giuseppe Lauria, Federico Verde, Isabella Fogh, Cinzia Tiloca, Giacomo P. Comi, Gianni Sorarù, Cristina Cereda, Philippe Corcia, Hannu Laaksovirta, Liisa Myllykangas, Lilja Jansson, Miko Valori, John Ealing, Hisham Hamdalla, Sara Rollinson, Stuart Pickering-Brown, Richard W. Orrell, Katie C. Sidle, Andrea Malaspina, John Hardy, Andrew B. Singleton, Janel O. Johnson, Sampath Arepalli, Peter C. Sapp, Diane McKenna-Yasek, Meraida Polak, Seneshaw Asress, Safa Al-Sarraj, Andrew King, Claire Troakes, Caroline Vance, Jacqueline de Belleroche, Frank Baas, Anneloor L.M.A. ten Asbroek, José Luis Muñoz-Blanco, Dena G. Hernandez, Jinhui Ding, J. Raphael Gibbs, Sonja W. Scholz, Mary Kay Floeter, Roy H. Campbell, Francesco Landi, Robert Bowser, Stefan M. Pulst, John M. Ravits, Daniel J.L. MacGowan, Janine Kirby, Erik P. Pioro, Roger Pamphlett, James Broach, Glenn Gerhard, Travis L. Dunckley, Christopher B. Brady, Neil W. Kowall, Juan C. Troncoso, Isabelle Le Ber, Kevin Mouzat, Serge Lumbroso, Terry D. Heiman-Patterson, Freya Kamel, Ludo Van Den Bosch, Robert H. Baloh, Tim M. Strom, Thomas Meitinger, Aleksey Shatunov, Kristel R. Van Eijk, Mamede de Carvalho, Maarten Kooyman, Bas Middelkoop, Matthieu Moisse, Russell L. McLaughlin, Michael A. Van Es, Markus Weber, Kevin B. Boylan, Marka Van Blitterswijk, Rosa Rademakers, Karen E. Morrison, A. Nazli Basak, Jesús S. Mora, Vivian E. Drory, Pamela J. Shaw, Martin R. Turner, Kevin Talbot, Orla Hardiman, Kelly L. Williams, Jennifer A. Fifita, Garth A. Nicholson, Ian P. Blair, Guy A. Rouleau, Jesús Esteban-Pérez, Alberto García-Redondo, Ammar Al-Chalabi, Ekaterina Rogaeva, Lorne Zinman, Lyle W. Ostrow, Nicholas J. Maragakis, Jeffrey D. Rothstein, Zachary Simmons, Johnathan Cooper-Knock, Alexis Brice, Stephen A. Goutman, Eva L. Feldman, Summer B. Gibson, Franco Taroni, Antonia Ratti, Cinzia Gellera, Philip Van Damme, Wim Robberecht, Pietro Fratta, Mario Sabatelli, Christian Lunetta, Albert C. Ludolph, Peter M. Andersen, Jochen H. Weishaupt, William Camu, John Q. Trojanowski, Vivianna M. Van Deerlin, Robert H. Brown, Leonard H. van den Berg, Jan H. Veldink, Matthew B. Harms, Jonathan D. Glass, David J. Stone, Pentti Tienari, Vincenzo Silani, Adriano Chiò, Christopher E. Shaw, Bryan J. Traynor, John E. Landers, Isabella Simone, Giancarlo Logroscino, Ilaria Bartolomei, Maria Rita Murru, Emanuela Costantino, Carla Pani, Roberta Puddu, Carla Caredda, Valeria Piras, Stefania Tranquilli, Stefania Cuccu, Daniela Corongiu, Maurizio Melis, Antonio Milia, Francesco Marrosu, Maria Giovanna Marrosu, Gianluca Floris, Antonino Cannas, Gianluigi Mancardi, Paola Origone, Paola Mandich, Sebastiano Cavallaro, Kalliopi Marinou, Riccardo Sideri, Silvana Penco, Lorena Mosca, Giuseppe Lauria Pinter, Massimo Corbo, Paola Carrera, Nicola Fini, Antonio Fasano, Lucio Tremolizzo, Alessandro Arosio, Carlo Ferrarese, Gioacchino Tedeschi, Maria Rosaria Monsurrò, Giovanni Piccirillo, Cinzia Femiano, Anna Ticca, Enzo Ortu, Rossella Spataro, Tiziana Colletti, Marcella Zollino, Amelia Conte, Marco Luigetti, Serena Lattante, Marialuisa Santarelli, Antonio Petrucci, Maura Pugliatti, Angelo Pirisi, Leslie D. Parish, Patrizia Occhineri, Fabio Giannini, Claudia Ricci, Michele Benigni, Tea B. Cau, Daniela Loi, Cristina Moglia, Maura Brunetti, Marco Barberis, Gabriella Restagno, Federico Casale, Giuseppe Marrali, Giuseppe Fuda, Irene Ossola, Stefania Cammarosano, Antonio Canosa, Antonio Ilardi, Umberto Manera, Maurizio Grassano, Raffaella Tanel, Fabrizio Pisano, Neil A. Shneider, Stephen Goutman, Siddharthan Chandran, Suvankar Pal, George Manousakis, Stanley H. Appel, Ericka Simpson, Leo Wang, Summer Gibson, Richard Bedlack, David Lacomis, Dhruv Sareen, Alexander Sherman, Lucie Bruijn, Michelle Penny, Andrew S. Allen, Stanley Appel, Richard S. Bedlack, Braden E. Boone, Robert Brown, John P. Carulli, Alessandra Chesi, Wendy K. Chung, Elizabeth T. Cirulli, Gregory M. Cooper, Julien Couthouis, Aaron G. Day-Williams, Patrick A. Dion, Yujun Han, Sebastian D. Hayes, Angela L. Jones, Jonathan Keebler, Brian J. Krueger, Brittany N. Lasseigne, Shawn E. Levy, Yi-Fan Lu, Tom Maniatis, Slavé Petrovski, Alya R. Raphael, Zhong Ren, Katherine B. Sims, John F. Staropoli, Lindsay L. Waite, Quanli Wang, Jack R. Wimbish, Winnie W. Xin, Justin Kwan, James R. Broach, Ximena Arcila-Londono, Edward B. Lee, Noah Zaitlen, Gregory A. Cox, Steve Finkbeiner, Efthimios Dardiotis, Eran Hornstein, Daniel J. MacGowan, Terry Heiman-Patterson, Molly G. Hammell, Nikolaos A. Patsopoulos, Joshua Dubnau, Avindra Nath, Stacia Wyman, Alexander LeNail, Jenny Van Eyk, Stephan Züchner, Rebecca Schule, Jacob McCauley, Sumaira Hussain, Anne Cooley, Marielle Wallace, Christine Clayman, Richard Barohn, Jeffrey Statland, John Ravits, Andrea Swenson, Carlayne Jackson, Jaya Trivedi, Shaida Khan, Jonathan Katz, Liberty Jenkins, Ted Burns, Kelly Gwathmey, James Caress, Corey McMillan, Lauren Elman, Erik Pioro, Jeannine Heckmann, Yuen So, David Walk, Samuel Maiser, Jinghui Zhang, Fabiola De Marchi, Stefania Corti, Mauro Ceroni, Gabriele Siciliano, Massimiliano Filosto, Maurizio Inghilleri, Silvia Peverelli, Claudia Colombrita, Barbara Poletti, Luca Maderna, Roberto Del Bo, Stella Gagliardi, Giorgia Querin, Cinzia Bertolin, Viviana Pensato, Barbara Castellotti, Vincent Meininger, Gérard Besson, Emmeline Lagrange, Pierre Clavelou, Nathalie Guy, Philippe Couratier, Patrick Vourch, Véronique Danel, Emilien Bernard, Gwendal Lemasson, Ahmad Al Kheifat, Peter Andersen, Adriano Chio, Jonathan Cooper-Knock, Annelot Dekker, Vivian Drory, Alberto Garcia Redondo, Marc Gotkine, Winston Hide, Alfredo Iacoangeli, Jonathan Glass, Kevin Kenna, Matthew Kiernan, John Landers, Russell McLaughlin, Jonathan Mill, Miguel Mitne Neto, Mattieu Moisse, Jesus Mora Pardina, Karen Morrison, Stephen Newhouse, Susana Pinto, Sara Pulit, Pamela Shaw, Chris Shaw, William Sproviero, Gijs Tazelaar, Philip van Damme, Leonard van den Berg, Rick van der Spek, Kristel van Eijk, Michael van Es, Joke van Vugt, Jan Veldink, Mayana Zatz, Denis C. Bauer, Natalie A. Twine, Department of Neurosciences, Pentti Tienari / Principal Investigator, Neurologian yksikkö, Research Programs Unit, Clinicum, Research Programme for Molecular Neurology, University of Helsinki, Medicum, Department of Pathology, HUS Neurocenter, Nicolas A., Kenna K.P., Renton A.E., Ticozzi N., Faghri F., Chia R., Dominov J.A., Kenna B.J., Nalls M.A., Keagle P., Rivera A.M., van Rheenen W., Murphy N.A., van Vugt J.J.F.A., Geiger J.T., Van der Spek R.A., Pliner H.A., Shankaracharya, Smith B.N., Marangi G., Topp S.D., Abramzon Y., Gkazi A.S., Eicher J.D., Kenna A., Logullo F.O., Simone I.L., Logroscino G., Salvi F., Bartolomei I., Borghero G., Murru M.R., Costantino E., Pani C., Puddu R., Caredda C., Piras V., Tranquilli S., Cuccu S., Corongiu D., Melis M., Milia A., Marrosu F., Marrosu M.G., Floris G., Cannas A., Capasso M., Caponnetto C., Mancardi G., Origone P., Mandich P., Conforti F.L., Cavallaro S., Mora G., Marinou K., Sideri R., Penco S., Mosca L., Lunetta C., Pinter G.L., Corbo M., Riva N., Carrera P., Volanti P., Mandrioli J., Fini N., Fasano A., Tremolizzo L., Arosio A., Ferrarese C., Trojsi F., Tedeschi G., Monsurro M.R., Piccirillo G., Femiano C., Ticca A., Ortu E., La Bella V., Spataro R., Colletti T., Sabatelli M., Zollino M., Conte A., Luigetti M., Lattante S., Santarelli M., Petrucci A., Pugliatti M., Pirisi A., Parish L.D., Occhineri P., Giannini F., Battistini S., Ricci C., Benigni M., Cau T.B., Loi D., Calvo A., Moglia C., Brunetti M., Barberis M., Restagno G., Casale F., Marrali G., Fuda G., Ossola I., Cammarosano S., Canosa A., Ilardi A., Manera U., Grassano M., Tanel R., Pisano F., Mazzini L., Messina S., D'Alfonso S., Corrado L., Ferrucci L., Harms M.B., Goldstein D.B., Shneider N.A., Goutman S.A., Simmons Z., Miller T.M., Chandran S., Pal S., Manousakis G., Appel S.H., Simpson E., Wang L., Baloh R.H., Gibson S.B., Bedlack R., Lacomis D., Sareen D., Sherman A., Bruijn L., Penny M., Moreno C.D.A.M., Kamalakaran S., Allen A.S., Boone B.E., Brown R.H., Carulli J.P., Chesi A., Chung W.K., Cirulli E.T., Cooper G.M., Couthouis J., Day-Williams A.G., Dion P.A., Gitler A.D., Glass J.D., Han Y., Harris T., Hayes S.D., Jones A.L., Keebler J., Krueger B.J., Lasseigne B.N., Levy S.E., Lu Y.-F., Maniatis T., McKenna-Yasek D., Myers R.M., Petrovski S., Pulst S.M., Raphael A.R., Ravits J.M., Ren Z., Rouleau G.A., Sapp P.C., Sims K.B., Staropoli J.F., Waite L.L., Wang Q., Wimbish J.R., Xin W.W., Phatnani H., Kwan J., Broach J., Arcila-Londono X., Lee E.B., Van Deerlin V.M., Fraenkel E., Ostrow L.W., Baas F., Zaitlen N., Berry J.D., Malaspina A., Fratta P., Cox G.A., Thompson L.M., Finkbeiner S., Dardiotis E., Hornstein E., MacGowan D.J.L., Heiman-Patterson T., Hammell M.G., Patsopoulos N.A., Dubnau J., Nath A., Musunuri R.L., Evani U.S., Abhyankar A., Zody M.C., Kaye J., Wyman S.K., LeNail A., Lima L., Rothstein J.D., Svendsen C.N., Van Eyk J.E., Maragakis N.J., Kolb S.J., Cudkowicz M., Baxi E., Benatar M., Taylor J.P., Wu G., Rampersaud E., Wuu J., Rademakers R., Zuchner S., Schule R., McCauley J., Hussain S., Cooley A., Wallace M., Clayman C., Barohn R., Statland J., Swenson A., Jackson C., Trivedi J., Khan S., Katz J., Jenkins L., Burns T., Gwathmey K., Caress J., McMillan C., Elman L., Pioro E.P., Heckmann J., So Y., Walk D., Maiser S., Zhang J., Silani V., Gellera C., Ratti A., Taroni F., Lauria G., Verde F., Fogh I., Tiloca C., Comi G.P., Soraru G., Cereda C., De Marchi F., Corti S., Ceroni M., Siciliano G., Filosto M., Inghilleri M., Peverelli S., Colombrita C., Poletti B., Maderna L., Del Bo R., Gagliardi S., Querin G., Bertolin C., Pensato V., Castellotti B., Camu W., Mouzat K., Lumbroso S., Corcia P., Meininger V., Besson G., Lagrange E., Clavelou P., Guy N., Couratier P., Vourch P., Danel V., Bernard E., Lemasson G., Laaksovirta H., Myllykangas L., Jansson L., Valori M., Ealing J., Hamdalla H., Rollinson S., Pickering-Brown S., Orrell R.W., Sidle K.C., Hardy J., Singleton A.B., Johnson J.O., Arepalli S., Polak M., Asress S., Al-Sarraj S., King A., Troakes C., Vance C., de Belleroche J., ten Asbroek A.L.M.A., Munoz-Blanco J.L., Hernandez D.G., Ding J., Gibbs J.R., Scholz S.W., Floeter M.K., Campbell R.H., Landi F., Bowser R., Kirby J., Pamphlett R., Gerhard G., Dunckley T.L., Brady C.B., Kowall N.W., Troncoso J.C., Le Ber I., Heiman-Patterson T.D., Kamel F., Van Den Bosch L., Strom T.M., Meitinger T., Shatunov A., Van Eijk K.R., de Carvalho M., Kooyman M., Middelkoop B., Moisse M., McLaughlin R.L., Van Es M.A., Weber M., Boylan K.B., Van Blitterswijk M., Morrison K.E., Basak A.N., Mora J.S., Drory V.E., Shaw P.J., Turner M.R., Talbot K., Hardiman O., Williams K.L., Fifita J.A., Nicholson G.A., Blair I.P., Esteban-Perez J., Garcia-Redondo A., Al-Chalabi A., Al Kheifat A., Andersen P.M., Chio A., Cooper-Knock J., Dekker A., Redondo A.G., Gotkine M., Hide W., Iacoangeli A., Kiernan M., Landers J.E., Mill J., Neto M.M., Pardina J.M., Newhouse S., Pinto S., Pulit S., Robberecht W., Shaw C., Sproviero W., Tazelaar G., Van Damme P., van den Berg L.H., van Vugt J., Veldink J.H., Zatz M., Bauer D.C., Twine N.A., Rogaeva E., Zinman L., Brice A., Feldman E.L., Ludolph A.C., Weishaupt J.H., Trojanowski J.Q., Stone D.J., Tienari P., Shaw C.E., Traynor B.J., ITALSGEN Consortium, Genomic Translation ALS Care GTAC, ALS Sequencing Consortium, NYGC ALS Consortium, Answer ALS Fdn, Clinical Res ALS Related Disorders, SLAGEN Consortium, French ALS Consortium, Project MinE ALS Sequencing Consor, Medical Research Council (MRC), ANS - Complex Trait Genetics, Human Genetics, ARD - Amsterdam Reproduction and Development, Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven), Università cattolica del Sacro Cuore [Roma] (Unicatt), Centre référent Sclérose Latérale Amyotrophique [CHRU Montpellier] (SLA CHRU Montpellier), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier)-Université Montpellier 1 (UM1), Lunar and Planetary Laboratory [Tucson] (LPL), University of Arizona, Università degli studi di Torino (UNITO), Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Centre National de la Recherche Scientifique (CNRS), New York Genome Center [New York], New York Genome Center, Laboratoire de Biométrie et Biologie Evolutive - UMR 5558 (LBBE), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de Recherche en Informatique et en Automatique (Inria)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS), St Jude Children's Research Hospital, Howard Hughes Medical Institute [Chevy Chase] (HHMI), Howard Hughes Medical Institute (HHMI), Centre de compétence de la Sclérose Latérale Amyotrophique [CHRU Tours] (SLA CHRU Tours), Centre Hospitalier Régional Universitaire de Tours (CHRU Tours), University College of London [London] (UCL), Synchrotron SOLEIL (SSOLEIL), Centre National de la Recherche Scientifique (CNRS), King‘s College London, University of New Haven [Connecticut], Princeton University, Laboratoire de Biochimie [CHRU Nîmes], Centre Hospitalier Universitaire de Nîmes (CHU Nîmes), Institut des Neurosciences de Montpellier - Déficits sensoriels et moteurs (INM), Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), Helmholtz-Zentrum München (HZM), University Medical Center [Utrecht], Deutsches Forschungszentrum für Künstliche Intelligenz GmbH = German Research Center for Artificial Intelligence (DFKI), Mayo Clinic [Jacksonville], Trinity College Dublin, Maurice Wohl Clinical Neuroscience Institut, Tanz Center Research in Neurodegenerative Diseases [Toronto], University of Toronto, Neurologie et thérapeutique expérimentale, Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR70-Université Pierre et Marie Curie - Paris 6 (UPMC), Repositório da Universidade de Lisboa, Nicolas, A, Kenna, K, Renton, A, Ticozzi, N, Faghri, F, Chia, R, Dominov, J, Kenna, B, Nalls, M, Keagle, P, Rivera, A, van Rheenen, W, Murphy, N, van Vugt, J, Geiger, J, van der Spek, R, Pliner, H, Shankaracharya, N, Smith, B, Marangi, G, Topp, S, Abramzon, Y, Gkazi, A, Eicher, J, Kenna, A, Logullo, F, Simone, I, Logroscino, G, Salvi, F, Bartolomei, I, Borghero, G, Murru, M, Costantino, E, Pani, C, Puddu, R, Caredda, C, Piras, V, Tranquilli, S, Cuccu, S, Corongiu, D, Melis, M, Milia, A, Marrosu, F, Marrosu, M, Floris, G, Cannas, A, Capasso, M, Caponnetto, C, Mancardi, G, Origone, P, Mandich, P, Conforti, F, Cavallaro, S, Mora, G, Marinou, K, Sideri, R, Penco, S, Mosca, L, Lunetta, C, Pinter, G, Corbo, M, Riva, N, Carrera, P, Volanti, P, Mandrioli, J, Fini, N, Fasano, A, Tremolizzo, L, Arosio, A, Ferrarese, C, Trojsi, F, Tedeschi, G, Monsurrò, M, Piccirillo, G, Femiano, C, Ticca, A, Ortu, E, La Bella, V, Spataro, R, Colletti, T, Sabatelli, M, Zollino, M, Conte, A, Luigetti, M, Lattante, S, Santarelli, M, Petrucci, A, Pugliatti, M, Pirisi, A, Parish, L, Occhineri, P, Giannini, F, Battistini, S, Ricci, C, Benigni, M, Cau, T, Loi, D, Calvo, A, Moglia, C, Brunetti, M, Barberis, M, Restagno, G, Casale, F, Marrali, G, Fuda, G, Ossola, I, Cammarosano, S, Canosa, A, Ilardi, A, Manera, U, Grassano, M, Tanel, R, Pisano, F, Mazzini, L, Messina, S, D'Alfonso, S, Corrado, L, Ferrucci, L, Harms, M, Goldstein, D, Shneider, N, Goutman, S, Simmons, Z, Miller, T, Chandran, S, Pal, S, Manousakis, G, Appel, S, Simpson, E, Wang, L, Baloh, R, Gibson, S, Bedlack, R, Lacomis, D, Sareen, D, Sherman, A, Bruijn, L, Penny, M, Moreno, C, Kamalakaran, S, Allen, A, Boone, B, Brown, R, Carulli, J, Chesi, A, Chung, W, Cirulli, E, Cooper, G, Couthouis, J, Day-Williams, A, Dion, P, Gitler, A, Glass, J, Han, Y, Harris, T, Hayes, S, Jones, A, Keebler, J, Krueger, B, Lasseigne, B, Levy, S, Lu, Y, Maniatis, T, McKenna-Yasek, D, Myers, R, Petrovski, S, Pulst, S, Raphael, A, Ravits, J, Ren, Z, Rouleau, G, Sapp, P, Sims, K, Staropoli, J, Waite, L, Wang, Q, Wimbish, J, Xin, W, Phatnani, H, Kwan, J, Broach, J, Arcila-Londono, X, Lee, E, Van Deerlin, V, Fraenkel, E, Ostrow, L, Baas, F, Zaitlen, N, Berry, J, Malaspina, A, Fratta, P, Cox, G, Thompson, L, Finkbeiner, S, Dardiotis, E, Hornstein, E, Macgowan, D, Heiman-Patterson, T, Hammell, M, Patsopoulos, N, Dubnau, J, Nath, A, Musunuri, R, Evani, U, Abhyankar, A, Zody, M, Kaye, J, Wyman, S, Lenail, A, Lima, L, Rothstein, J, Svendsen, C, Van Eyk, J, Maragakis, N, Kolb, S, Cudkowicz, M, Baxi, E, Benatar, M, Taylor, J, Wu, G, Rampersaud, E, Wuu, J, Rademakers, R, Züchner, S, Schule, R, Mccauley, J, Hussain, S, Cooley, A, Wallace, M, Clayman, C, Barohn, R, Statland, J, Swenson, A, Jackson, C, Trivedi, J, Khan, S, Katz, J, Jenkins, L, Burns, T, Gwathmey, K, Caress, J, Mcmillan, C, Elman, L, Pioro, E, Heckmann, J, So, Y, Walk, D, Maiser, S, Zhang, J, Silani, V, Gellera, C, Ratti, A, Taroni, F, Lauria, G, Verde, F, Fogh, I, Tiloca, C, Comi, G, Sorarù, G, Cereda, C, De Marchi, F, Corti, S, Ceroni, M, Siciliano, G, Filosto, M, Inghilleri, M, Peverelli, S, Colombrita, C, Poletti, B, Maderna, L, Del Bo, R, Gagliardi, S, Querin, G, Bertolin, C, Pensato, V, Castellotti, B, Camu, W, Mouzat, K, Lumbroso, S, Corcia, P, Meininger, V, Besson, G, Lagrange, E, Clavelou, P, Guy, N, Couratier, P, Vourch, P, Danel, V, Bernard, E, Lemasson, G, Laaksovirta, H, Myllykangas, L, Jansson, L, Valori, M, Ealing, J, Hamdalla, H, Rollinson, S, Pickering-Brown, S, Orrell, R, Sidle, K, Hardy, J, Singleton, A, Johnson, J, Arepalli, S, Polak, M, Asress, S, Al-Sarraj, S, King, A, Troakes, C, Vance, C, de Belleroche, J, ten Asbroek, A, Muñoz-Blanco, J, Hernandez, D, Ding, J, Gibbs, J, Scholz, S, Floeter, M, Campbell, R, Landi, F, Bowser, R, Kirby, J, Pamphlett, R, Gerhard, G, Dunckley, T, Brady, C, Kowall, N, Troncoso, J, Le Ber, I, Kamel, F, Van Den Bosch, L, Strom, T, Meitinger, T, Shatunov, A, Van Eijk, K, de Carvalho, M, Kooyman, M, Middelkoop, B, Moisse, M, Mclaughlin, R, Van Es, M, Weber, M, Boylan, K, Van Blitterswijk, M, Morrison, K, Basak, A, Mora, J, Drory, V, Shaw, P, Turner, M, Talbot, K, Hardiman, O, Williams, K, Fifita, J, Nicholson, G, Blair, I, Esteban-Pérez, J, García-Redondo, A, Al-Chalabi, A, Al Kheifat, A, Andersen, P, Chio, A, Cooper-Knock, J, Dekker, A, Redondo, A, Gotkine, M, Hide, W, Iacoangeli, A, Kiernan, M, Landers, J, Mill, J, Neto, M, Pardina, J, Newhouse, S, Pinto, S, Pulit, S, Robberecht, W, Shaw, C, Sproviero, W, Tazelaar, G, van Damme, P, van den Berg, L, van Eijk, K, van Es, M, Veldink, J, Zatz, M, Bauer, D, Twine, N, Rogaeva, E, Zinman, L, Brice, A, Feldman, E, Ludolph, A, Weishaupt, J, Trojanowski, J, Stone, D, Tienari, P, Chiò, A, Traynor, B, Nicolas, Aude, Kenna, Kevin P, Renton, Alan E, Ticozzi, Nicola, Faghri, Faraz, Chia, Ruth, Dominov, Janice A, Kenna, Brendan J, Nalls, Mike A, Keagle, Pamela, Rivera, Alberto M, van Rheenen, Wouter, Murphy, Natalie A, van Vugt, Joke J F A, Geiger, Joshua T, Van der Spek, Rick A, Pliner, Hannah A, Shankaracharya, Null, Smith, Bradley N, Marangi, Giuseppe, Topp, Simon D, Abramzon, Yevgeniya, Gkazi, Athina Soragia, Eicher, John D, Kenna, Aoife, Mora, Gabriele, Calvo, Andrea, Mazzini, Letizia, Riva, Nilo, Mandrioli, Jessica, Caponnetto, Claudia, Battistini, Stefania, Volanti, Paolo, La Bella, Vincenzo, Conforti, Francesca L, Borghero, Giuseppe, Messina, Sonia, Simone, Isabella L, Trojsi, Francesca, Salvi, Fabrizio, Logullo, Francesco O, D'Alfonso, Sandra, Corrado, Lucia, Capasso, Margherita, Ferrucci, Luigi, Logullo, Fo, Murru, Mr, Marrosu, Mg, Conforti, Fl, Pinter, Gl, Tedeschi, Gioacchino, Monsurrò, Maria Rosaria, Parish, Ld, Cau, Tb, Moreno, Cristiane de Araujo Martin, Kamalakaran, Sitharthan, Goldstein, David B, Gitler, Aaron D, Harris, Tim, Myers, Richard M, Phatnani, Hemali, Musunuri, Rajeeva Lochan, Evani, Uday Shankar, Abhyankar, Avinash, Zody, Michael C, Kaye, Julia, Finkbeiner, Steven, Wyman, Stacia K, Lenail, Alex, Lima, Leandro, Fraenkel, Ernest, Svendsen, Clive N, Thompson, Leslie M, Van Eyk, Jennifer E, Berry, James D, Miller, Timothy M, Kolb, Stephen J, Cudkowicz, Merit, Baxi, Emily, Benatar, Michael, Taylor, J Paul, Rampersaud, Evadnie, Wu, Gang, Wuu, Joanne, Lauria, Giuseppe, Verde, Federico, Fogh, Isabella, Tiloca, Cinzia, Comi, Giacomo P, Sorarù, Gianni, Cereda, Cristina, Corcia, Philippe, Laaksovirta, Hannu, Myllykangas, Liisa, Jansson, Lilja, Valori, Miko, Ealing, John, Hamdalla, Hisham, Rollinson, Sara, Pickering-Brown, Stuart, Orrell, Richard W, Sidle, Katie C, Malaspina, Andrea, Hardy, John, Singleton, Andrew B, Johnson, Janel O, Arepalli, Sampath, Sapp, Peter C, McKenna-Yasek, Diane, Polak, Meraida, Asress, Seneshaw, Al-Sarraj, Safa, King, Andrew, Troakes, Claire, Vance, Caroline, de Belleroche, Jacqueline, Baas, Frank, Ten Asbroek, Anneloor L M A, Muñoz-Blanco, José Lui, Hernandez, Dena G, Ding, Jinhui, Gibbs, J Raphael, Scholz, Sonja W, Floeter, Mary Kay, Campbell, Roy H, Landi, Francesco, Bowser, Robert, Pulst, Stefan M, Ravits, John M, Macgowan, Daniel J L, Kirby, Janine, Pioro, Erik P, Pamphlett, Roger, Broach, Jame, Gerhard, Glenn, Dunckley, Travis L, Brady, Christopher B, Kowall, Neil W, Troncoso, Juan C, Le Ber, Isabelle, Mouzat, Kevin, Lumbroso, Serge, Heiman-Patterson, Terry D, Kamel, Freya, Van Den Bosch, Ludo, Baloh, Robert H, Strom, Tim M, Meitinger, Thoma, Shatunov, Aleksey, Van Eijk, Kristel R, de Carvalho, Mamede, Kooyman, Maarten, Middelkoop, Ba, Moisse, Matthieu, Mclaughlin, Russell L, Van Es, Michael A, Weber, Marku, Boylan, Kevin B, Van Blitterswijk, Marka, Rademakers, Rosa, Morrison, Karen E, Basak, A Nazli, Mora, Jesús S, Drory, Vivian E, Shaw, Pamela J, Turner, Martin R, Talbot, Kevin, Hardiman, Orla, Williams, Kelly L, Fifita, Jennifer A, Nicholson, Garth A, Blair, Ian P, Rouleau, Guy A, Esteban-Pérez, Jesú, García-Redondo, Alberto, Al-Chalabi, Ammar, Rogaeva, Ekaterina, Zinman, Lorne, Ostrow, Lyle W, Maragakis, Nicholas J, Rothstein, Jeffrey D, Simmons, Zachary, Cooper-Knock, Johnathan, Brice, Alexi, Goutman, Stephen A, Feldman, Eva L, Gibson, Summer B, Taroni, Franco, Ratti, Antonia, Gellera, Cinzia, Van Damme, Philip, Robberecht, Wim, Fratta, Pietro, Sabatelli, Mario, Lunetta, Christian, Ludolph, Albert C, Andersen, Peter M, Weishaupt, Jochen H, Camu, William, Trojanowski, John Q, Van Deerlin, Vivianna M, Brown, Robert H, van den Berg, Leonard H, Veldink, Jan H, Harms, Matthew B, Glass, Jonathan D, Stone, David J, Tienari, Pentti, Silani, Vincenzo, Chiò, Adriano, Shaw, Christopher E, Traynor, Bryan J, Landers, John E, Université Montpellier 1 (UM1)-Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Régional Universitaire de Tours (CHRU TOURS), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
Male ,Als gene ,Genome-wide association study ,FAMILIAL ALS ,ALS ,axonal transport ,cargo ,GWAS ,KIF5A ,WES ,WGS ,0302 clinical medicine ,80 and over ,Psychology ,Aetiology ,Aged, 80 and over ,0303 health sciences ,French ALS Consortium ,Kinesin ,KINESIN HEAVY-CHAIN ,Cognitive Sciences ,Human ,Hereditary spastic paraplegia ,Neuroscience(all) ,Single-nucleotide polymorphism ,TARGETED DISRUPTION ,Article ,03 medical and health sciences ,Genetics ,Humans ,Amino Acid Sequence ,Loss function ,Aged ,HEXANUCLEOTIDE REPEAT ,Neuroscience (all) ,MUTATIONS ,Amyotrophic Lateral Sclerosis ,3112 Neurosciences ,1702 Cognitive Science ,medicine.disease ,ITALSGEN Consortium ,Answer ALS Foundation ,030104 developmental biology ,ALS Sequencing Consortium ,Human medicine ,1109 Neurosciences ,030217 neurology & neurosurgery ,0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Kinesins ,Neurodegenerative ,Genetic analysis ,Genome ,AMYOTROPHIC-LATERAL-SCLEROSIS ,3124 Neurology and psychiatry ,Cohort Studies ,Pathogenesis ,Loss of Function Mutation ,Missense mutation ,2.1 Biological and endogenous factors ,Amyotrophic lateral sclerosis ,NYGC ALS Consortium ,General Neuroscience ,ALS, axonal transport, cargo, GWAS, KIF5A, WES, WGS ,Middle Aged ,Phenotype ,Settore MED/26 - NEUROLOGIA ,Neurological ,Project MinE ALS Sequencing Consortium ,Female ,Adult ,Biology ,GENOTYPE IMPUTATION ,Genome-Wide Association Study ,Young Adult ,NO ,Rare Diseases ,medicine ,SLAGEN Consortium ,Gene ,030304 developmental biology ,Clinical Research in ALS and Related Disorders for Therapeutic Development (CReATe) Consortium ,Neurology & Neurosurgery ,Human Genome ,Neurosciences ,AXONAL-TRANSPORT ,Brain Disorders ,Family member ,DNA-DAMAGE ,MOTOR-NEURONS ,3111 Biomedicine ,Cohort Studie ,Genomic Translation for ALS Care (GTAC) Consortium ,Amyotrophic Lateral Sclerosi - Abstract
© 2018 Elsevier Inc., To identify novel genes associated with ALS, we undertook two lines of investigation. We carried out a genome-wide association study comparing 20,806 ALS cases and 59,804 controls. Independently, we performed a rare variant burden analysis comparing 1,138 index familial ALS cases and 19,494 controls. Through both approaches, we identified kinesin family member 5A (KIF5A) as a novel gene associated with ALS. Interestingly, mutations predominantly in the N-terminal motor domain of KIF5A are causative for two neurodegenerative diseases: hereditary spastic paraplegia (SPG10) and Charcot-Marie-Tooth type 2 (CMT2). In contrast, ALS-associated mutations are primarily located at the C-terminal cargo-binding tail domain and patients harboring loss-of-function mutations displayed an extended survival relative to typical ALS cases. Taken together, these results broaden the phenotype spectrum resulting from mutations in KIF5A and strengthen the role of cytoskeletal defects in the pathogenesis of ALS.
- Published
- 2018
- Full Text
- View/download PDF
18. Mutational analysis of TARDBP in neurodegenerative diseases
- Author
-
Ashley Lyn Leclerc, Pamela Keagle, Marka van Blitterswijk, Nicola Ticozzi, Robert H. Brown, Diane McKenna-Yasek, John Landers, Vincenzo Silani, Anne-Marie Wills, and Peter C. Sapp
- Subjects
Aging ,DNA Mutational Analysis ,medicine.disease_cause ,TARDBP ,Article ,Pathogenesis ,Exon ,Alzheimer Disease ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Genetics ,Mutation ,business.industry ,General Neuroscience ,Amyotrophic Lateral Sclerosis ,Parkinson Disease ,Exons ,medicine.disease ,DNA-Binding Proteins ,Mutational analysis ,Neurology (clinical) ,Geriatrics and Gerontology ,Alzheimer's disease ,business ,Developmental Biology - Abstract
Neurodegenerative diseases are often characterized by the presence of aggregates of misfolded proteins. TDP-43 is a major component of these aggregates in amyotrophic lateral sclerosis (ALS), but has also been observed in Alzheimer's (AD) and Parkinson's Diseases (PD). In addition, mutations in the TARDBP gene, encoding TDP-43, have been found to be a significant cause of familial ALS (FALS). All mutations, except for one, have been found in exon 6. To confirm this observation in ALS and to investigate whether TARDBP may play a role in the pathogenesis of AD and PD, we screened for mutations in exon 6 of the TARDBP gene in three cohorts composed of 376 AD, 463 PD (18% familial PD) and 376 ALS patients (50% FALS). We found mutations in ∼ 7% of FALS and ∼0.5% of sporadic ALS (SALS) patients, including two novel mutations, p.N352T and p.G384R. In contrast, we did not find TARDBP mutations in our cohort of AD and PD patients. These results suggest that mutations in TARDBP are not a significant cause of AD and PD.
- Published
- 2011
- Full Text
- View/download PDF
19. Mutational analysis reveals the FUS homolog TAF15 as a candidate gene for familial amyotrophic lateral sclerosis
- Author
-
Vincenzo Silani, Daryl A. Bosco, Nicola Ticozzi, John Landers, Jonathan D. Glass, Christopher Shaw, Robert H. Brown, Peter C. Sapp, Diane McKenna-Yasek, Caroline Vance, Ashley Lyn Leclerc, and Pamela Keagle
- Subjects
Candidate gene ,DNA Mutational Analysis ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,TARDBP ,Cellular and Molecular Neuroscience ,medicine ,Humans ,Missense mutation ,Amino Acid Sequence ,Amyotrophic lateral sclerosis ,Gene ,Genetic Association Studies ,Genetics (clinical) ,TAF15 ,Genetics ,TATA-Binding Protein Associated Factors ,Mutation ,Base Sequence ,Sequence Homology, Amino Acid ,Amyotrophic Lateral Sclerosis ,Genetic Variation ,medicine.disease ,Psychiatry and Mental health ,RNA-Binding Protein FUS - Abstract
FUS, EWS, and TAF15 belong to the TET family of structurally similar DNA/RNA-binding proteins. Mutations in the FUS gene have recently been discovered as a cause of familial amyotrophic lateral sclerosis (FALS). Given the structural and functional similarities between the three genes, we screened TAF15 and EWS in 263 and 94 index FALS cases, respectively. No coding variants were found in EWS, while we identified six novel changes in TAF15. Of these, two 24 bp deletions and a R388H missense variant were also found in healthy controls. A D386N substitution was shown not to segregate with the disease in the affected pedigree. A single A31T and two R395Q changes were identified in FALS cases but not in over 1,100 controls. Interestingly, one of the R395Q FALS cases also harbors a TARDBP mutation (G384R). Altogether, these results suggest that additional studies are needed to determine whether mutations in the TAF15 gene represent a cause of FALS.
- Published
- 2011
- Full Text
- View/download PDF
20. Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS
- Author
-
Yuyu Song, Nathan Lemay, François Gros-Louis, Robert H. Brown, Matthew P. Frosch, Jeffrey N. Agar, Daryl A. Bosco, Holly Goolsby, Benjamin A. Fontaine, Piera Pasinelli, Gerardo Morfini, Jean-Pierre Julien, N. Murat Karabacak, Diane McKenna-Yasek, and Scott T. Brady
- Subjects
Adult ,Male ,Models, Molecular ,Protein Folding ,animal diseases ,SOD1 ,Crystallography, X-Ray ,medicine.disease_cause ,Mass Spectrometry ,Article ,Superoxide dismutase ,03 medical and health sciences ,Superoxide Dismutase-1 ,0302 clinical medicine ,parasitic diseases ,medicine ,Humans ,Proteostasis Deficiencies ,Amyotrophic lateral sclerosis ,Aged ,030304 developmental biology ,0303 health sciences ,Mutation ,biology ,Superoxide Dismutase ,General Neuroscience ,Amyotrophic Lateral Sclerosis ,Wild type ,nutritional and metabolic diseases ,Middle Aged ,Motor neuron ,medicine.disease ,Molecular biology ,Antibodies, Anti-Idiotypic ,nervous system diseases ,medicine.anatomical_structure ,Biochemistry ,nervous system ,biology.protein ,Kinesin ,Female ,Oxidation-Reduction ,Neuroscience ,Epitope Mapping ,030217 neurology & neurosurgery ,Conformational epitope - Abstract
Many mutations confer one or more toxic function(s) on copper/zinc superoxide dismutase 1 (SOD1) that impair motor neuron viability and cause familial amyotrophic lateral sclerosis (FALS). Using a conformation-specific antibody that detects misfolded SOD1 (C4F6), we found that oxidized wild-type SOD1 and mutant SOD1 share a conformational epitope that is not present in normal wild-type SOD1. In a subset of human sporadic ALS (SALS) cases, motor neurons in the lumbosacral spinal cord were markedly C4F6 immunoreactive, indicating that an aberrant wild-type SOD1 species was present. Recombinant, oxidized wild-type SOD1 and wild-type SOD1 immunopurified from SALS tissues inhibited kinesin-based fast axonal transport in a manner similar to that of FALS-linked mutant SOD1. Our findings suggest that wild-type SOD1 can be pathogenic in SALS and identify an SOD1-dependent pathogenic mechanism common to FALS and SALS.
- Published
- 2010
21. Mutant FUS proteins that cause amyotrophic lateral sclerosis incorporate into stress granules
- Author
-
Nathan Lemay, Hongru Zhou, Diane McKenna-Yasek, Robert H. Brown, Hae Kyung Ko, Daryl A. Bosco, Christopher J. Burke, Lawrence J. Hayward, Peter C. Sapp, and Thomas J. Kwiatkowski
- Subjects
Adult ,Male ,Cytoplasm ,Green Fluorescent Proteins ,Mutant ,Mutation, Missense ,Biology ,medicine.disease_cause ,Cell Line ,Stress granule ,Genetics ,medicine ,Animals ,Humans ,Missense mutation ,Heat shock ,Amyotrophic lateral sclerosis ,Molecular Biology ,Zebrafish ,Genetics (clinical) ,Mutation ,Amyotrophic Lateral Sclerosis ,Articles ,General Medicine ,Anatomy ,Middle Aged ,medicine.disease ,biology.organism_classification ,Cell biology ,Oxidative Stress ,RNA-Binding Protein FUS ,Female - Abstract
Mutations in the RNA-binding protein FUS (fused in sarcoma) are linked to amyotrophic lateral sclerosis (ALS), but the mechanism by which these mutants cause motor neuron degeneration is not known. We report a novel ALS truncation mutant (R495X) that leads to a relatively severe ALS clinical phenotype compared with FUS missense mutations. Expression of R495X FUS, which abrogates a putative nuclear localization signal at the C-terminus of FUS, in HEK-293 cells and in the zebrafish spinal cord caused a striking cytoplasmic accumulation of the protein to a greater extent than that observed for recessive (H517Q) and dominant (R521G) missense mutants. Furthermore, in response to oxidative stress or heat shock conditions in cultures and in vivo, the ALS-linked FUS mutants, but not wild-type FUS, assembled into perinuclear stress granules in proportion to their cytoplasmic expression levels. These findings demonstrate a potential link between FUS mutations and cellular pathways involved in stress responses that may be relevant to altered motor neuron homeostasis in ALS.
- Published
- 2010
- Full Text
- View/download PDF
22. Reduced expression of the Kinesin-Associated Protein 3 ( KIFAP3 ) gene increases survival in sporadic amyotrophic lateral sclerosis
- Author
-
Ammar Al-Chalabi, Ting Jan Cho, Thomas J. Kwiatkowski, Michael A. van Es, Hylke M. Blauw, H. Robert Horvitz, Christopher Shaw, Alayna Barnes-Nessa, Peter C. Sapp, Nicole R. Couture, Christiaan G J Saris, Roel A. Ophoff, Philippe Corcia, Betsy A. Hosler, Lijia Shi, Vincenzo Silani, Aslihan Ozoguz, Adrian J. Ivinson, François Salachas, Pilar Galan, Valerie K. Hansen, Robert H. Brown, John Powell, Orla Hardiman, Claire L. Simpson, Jonathan D. Glass, Diane McKenna-Yasek, Shaun Purcell, John Landers, Jan H. Veldink, Simon Cronin, Franck Georges, Nicola Ticozzi, P. Nigel Leigh, Paul W.J. van Vught, Vincent Meininger, John H. J. Wokke, Wendy J. Broom, Meraida Polak, Mark Lathrop, Simon Heath, Anne-Marie Wills, Ildefonso Rodriguez-Leyva, Leonard H. van den Berg, Frank P. Diekstra, and Judith Melki
- Subjects
Genetics ,Linkage disequilibrium ,Multidisciplinary ,Amyotrophic Lateral Sclerosis ,Single-nucleotide polymorphism ,Genome-wide association study ,Biological Sciences ,Biology ,medicine.disease ,Polymorphism, Single Nucleotide ,Cytoskeletal Proteins ,KIFAP3 ,medicine ,Humans ,SNP ,Allele ,Amyotrophic lateral sclerosis ,Promoter Regions, Genetic ,Alleles ,Adaptor Proteins, Signal Transducing ,SNP array - Abstract
Amyotrophic lateral sclerosis is a degenerative disorder of motor neurons that typically develops in the 6th decade and is uniformly fatal, usually within 5 years. To identify genetic variants associated with susceptibility and phenotypes in sporadic ALS, we performed a genome-wide SNP analysis in sporadic ALS cases and controls. A total of 288,357 SNPs were screened in a set of 1,821 sporadic ALS cases and 2,258 controls from the U.S. and Europe. Survival analysis was performed using 1,014 deceased sporadic cases. Top results for susceptibility were further screened in an independent sample set of 538 ALS cases and 556 controls. SNP rs1541160 within the KIFAP3 gene (encoding a kinesin-associated protein) yielded a genome-wide significant result ( P = 1.84 × 10 −8 ) that withstood Bonferroni correction for association with survival. Homozygosity for the favorable allele (CC) conferred a 14.0 months survival advantage. Sequence, genotypic and functional analyses revealed that there is linkage disequilibrium between rs1541160 and SNP rs522444 within the KIFAP3 promoter and that the favorable alleles of rs1541160 and rs522444 correlate with reduced KIFAP3 expression. No SNPs were associated with risk of sporadic ALS, site of onset, or age of onset. We have identified a variant within the KIFAP3 gene that is associated with decreased KIFAP3 expression and increased survival in sporadic ALS. These findings support the view that genetic factors modify phenotypes in this disease and that cellular motor proteins are determinants of motor neuron viability.
- Published
- 2009
- Full Text
- View/download PDF
23. Deleterious Variants of FIG4, a Phosphoinositide Phosphatase, in Patients with ALS
- Author
-
Adrienne E. Grant, Guy M. Lenk, Sarah K. Bergren, Peter C. Sapp, Robert H. Brown, Diane McKenna-Yasek, John Landers, Lois S. Weisman, Lesley Everett, Clement Y. Chow, Miriam H. Meisler, Julie M. Jones, Denise A. Figlewicz, Massachusetts Institute of Technology. Department of Biology, and Sapp, Peter C.
- Subjects
Nonsynonymous substitution ,Adult ,Heterozygote ,Phosphatase ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Report ,medicine ,Genetics ,Humans ,Genetic Predisposition to Disease ,Genetics(clinical) ,Amino Acid Sequence ,Amyotrophic lateral sclerosis ,Allele ,Motor Neuron Disease ,Genetics (clinical) ,030304 developmental biology ,Primary Lateral Sclerosis ,Aged ,0303 health sciences ,Mutation ,Flavoproteins ,Amyotrophic Lateral Sclerosis ,Heterozygote advantage ,Middle Aged ,medicine.disease ,Phosphoric Monoester Hydrolases ,3. Good health ,030217 neurology & neurosurgery - Abstract
Mutations of the lipid phosphatase FIG4 that regulates PI(3,5)P2 are responsible for the recessive peripheral-nerve disorder CMT4J. We now describe nonsynonymous variants of FIG4 in 2% (9/473) of patients with amyotrophic lateral sclerosis (ALS) and primary lateral sclerosis (PLS). Heterozygosity for a deleterious allele of FIG4 appears to be a risk factor for ALS and PLS, extending the list of known ALS genes and increasing the clinical spectrum of FIG4-related diseases., Howard Hughes Medical Institute (Investigator), National Institutes of Health (U.S.) (grant GM24872), National Institutes of Health (U.S.) (grant NS050557), National Institutes of Health (U.S.) (grant NIH NS050557), National Institutes of Health (U.S.) (grant NIH NS050641), National Institutes of Health (U.S.) (grant NIH T32 GM007544), Pierre L. de Bourgknecht ALS Research Foundation, Project ALS Foundation, Angel Fund
- Published
- 2009
- Full Text
- View/download PDF
24. A common haplotype within the PON1 promoter region is associated with sporadic ALS
- Author
-
Anne-Marie Wills, Ting-Jan Cho, Peter C. Sapp, John Landers, Diane McKenna-Yasek, Jonathan D. Glass, Lijia Shi, Ammar Al-Chalabi, Frank P. Diekstra, Christopher Shaw, Stephan Niemann, Meraida Polak, Ildefonso Rodriguez-Leyva, Bryan J. Traynor, P. Nigel Leigh, and Robert H. Brown
- Subjects
haplotypes ,Linkage disequilibrium ,Genotype ,SNP ,Single-nucleotide polymorphism ,Polymorphism, Single Nucleotide ,Linkage Disequilibrium ,Gene cluster ,medicine ,Humans ,Genetic Predisposition to Disease ,Amyotrophic lateral sclerosis ,Promoter Regions, Genetic ,Genetics ,biology ,Aryldialkylphosphatase ,case-control studies ,Amyotrophic Lateral Sclerosis ,Haplotype ,Paraoxonase ,Sequence Analysis, DNA ,General Medicine ,medicine.disease ,PON1 ,paraoxonase ,Isoenzymes ,Neurology ,Multigene Family ,biology.protein ,Original Article ,Neurology (clinical) ,Age of onset - Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, neurodegenerative disorder of upper and lower motor neurons. Genetic variants in the paraoxonase gene cluster have been associated with susceptibility to sporadic ALS. Because these studies have yielded conflicting results, we have further investigated this association in a larger data set. Twenty SNPs spanning the paraoxonase gene cluster were genotyped on a panel of 597 case and 692 control samples and tested for association with risk of sporadic ALS and with ALS sub-phenotypes. Our study revealed two SNPs, rs987539 and rs2074351, within the paraoxonase gene cluster that are associated with susceptibility to sporadic ALS (uncorrected p=6.47E-04 and 7.87E-04, respectively). None of the 20 SNPs displayed significant associations with age of onset, site of onset or disease survival. Using a sliding window approach, we have also identified a 5-SNP haplotype that is significantly associated with risk of sporadic ALS (p=2.75E-05). We conclude that a common haplotype within the PON1 promoter region is associated with susceptibility to sporadic ALS.
- Published
- 2008
- Full Text
- View/download PDF
25. 50bp deletion in the promoter for superoxide dismutase 1 (SOD1) reduces SOD1 expression in vitro and may correlate with increased age of onset of sporadic amyotrophic lateral sclerosis
- Author
-
Matthew Greenway, Caroline Hayward, Kelly E. Glajch, Robert J. Swingler, Ghazaleh Sadri-Vakili, Jonathan D. Glass, Wendy J. Broom, Paul N. Valdmanis, Vincent Meininger, Robert H. Brown, Carsten Russ, Peter C. Sapp, Nicolas Dupré, Kristen E. Auwarter, Jang-Ho J. Cha, Betsy A. Hosler, Orla Hardiman, Meraida Polack, Jean-Pierre Bouchard, Diane McKenna-Yasek, Shaun Purcell, and Guy A. Rouleau
- Subjects
Male ,Genotype ,Sp1 Transcription Factor ,DNA Mutational Analysis ,SOD1 ,Gene Expression ,Biology ,Superoxide Dismutase-1 ,Risk Factors ,Gene expression ,Humans ,Genetic Predisposition to Disease ,Age of Onset ,Promoter Regions, Genetic ,Gene ,Sequence Deletion ,Genetic association ,Genetics ,Reporter gene ,Polymorphism, Genetic ,Base Sequence ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,Homozygote ,Quebec ,General Medicine ,Middle Aged ,Molecular biology ,United States ,Phenotype ,Scotland ,Neurology ,Regulatory sequence ,Female ,Neurology (clinical) ,Age of onset ,Ireland ,Chromatin immunoprecipitation - Abstract
The objective was to test the hypothesis that a described association between homozygosity for a 50bp deletion in the SOD1 promoter 1684bp upstream of the SOD1 ATG and an increased age of onset in SALS can be replicated in additional SALS and control sample sets from other populations. Our second objective was to examine whether this deletion attenuates expression of the SOD1 gene. Genomic DNA from more than 1200 SALS cases from Ireland, Scotland, Quebec and the USA was genotyped for the 50bp SOD1 promoter deletion. Reporter gene expression analysis, electrophoretic mobility shift assays and chromatin immunoprecipitation studies were utilized to examine the functional effects of the deletion. The genetic association for homozygosity for the promoter deletion with an increased age of symptom onset was confirmed overall in this further study (p=0.032), although it was only statistically significant in the Irish subset, and remained highly significant in the combined set of all cohorts (p=0.001). Functional studies demonstrated that this polymorphism reduces the activity of the SOD1 promoter by approximately 50%. In addition we revealed that the transcription factor SP1 binds within the 50bp deletion region in vitro and in vivo. Our findings suggest the hypothesis that this deletion reduces expression of the SOD1 gene and that levels of the SOD1 protein may modify the phenotype of SALS within selected populations.
- Published
- 2008
- Full Text
- View/download PDF
26. Birth order and the genetics of amyotrophic lateral sclerosis
- Author
-
C. Johnston, Umesh Vivekananda, Diane McKenna-Yasek, Ammar Al-Chalabi, Peter Leigh, Robert H. Brown, and Christopher Shaw
- Subjects
Genetic Markers ,Male ,Aging ,medicine.risk_factor ,DNA Mutational Analysis ,Inheritance Patterns ,Pedigree chart ,Biology ,Degenerative disease ,Risk Factors ,medicine ,Humans ,Point Mutation ,Genetic Predisposition to Disease ,Family history ,Paternal age effect ,Amyotrophic lateral sclerosis ,Association mapping ,Retrospective Studies ,Genetics ,Polymorphism, Genetic ,Siblings ,Point mutation ,Amyotrophic Lateral Sclerosis ,medicine.disease ,Pedigree ,Birth order ,Neurology ,Female ,Neurology (clinical) ,Birth Order ,Gene Deletion - Abstract
The cause of ALS remains largely unknown for the 90% with no known family history, but spontaneous mutation to risk alleles of as yet unidentified genes is possible. It has long been recognized that genetic diseases may be more likely to occur in the last born children of a sibship because increased paternal age is associated with an increased spontaneous point mutation rate in sperm. To test the hypothesis that such a mechanism is responsible for sporadic ALS, we have performed a retrospective analysis of birth order position. We have analyzed sibships of size greater than four using a binomial test for birth position. The 478 pedigrees studied show no birth order effect, suggesting that any genetic contributions to sporadic ALS are more likely to be through deletion in large genes or interactions of common polymorphisms, rather than frequent spontaneous point mutation. This is encouraging for the prospect of finding sporadic ALS susceptibility genes using genome-wide association mapping.
- Published
- 2007
- Full Text
- View/download PDF
27. Failure to detect enterovirus in the spinal cord of ALS patients using a sensitive RT-PCR method
- Author
-
Raymond P. Roos, Diane McKenna-Yasek, M. S. Oberste, Robert H. Brown, M. M. Berger, William A. Nix, Mark A. Pallansch, and Benjamin Rix Brooks
- Subjects
Pathology ,medicine.medical_specialty ,Central nervous system ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Sensitivity and Specificity ,Virus ,Mice ,Freezing ,medicine ,Animals ,Humans ,False Positive Reactions ,Enterovirus ,Reverse Transcriptase Polymerase Chain Reaction ,Poliovirus ,Amyotrophic Lateral Sclerosis ,RNA ,Spinal cord ,Reverse transcriptase ,Real-time polymerase chain reaction ,medicine.anatomical_structure ,Spinal Cord ,RNA, Viral ,Capsid Proteins ,Neurology (clinical) ,5' Untranslated Regions - Abstract
Objective: To assess the association of enteroviruses (EV) with ALS by applying a sensitive seminested reverse transcription (RT) PCR protocol to the detection of enteroviral RNA in a blinded set of archived tissues from ALS and control cases.Methods: The specimen set consisted of 24 frozen spinal cord samples from ALS cases, 17 frozen spinal cord samples from negative control (non-ALS) cases, and 5 frozen spinal cord positive control samples. The positive controls were two human spinal cord samples spiked with poliovirus (PV) and three spinal cords from PV-infected transgenic mice. A sensitive, EV-specific, seminested RT-PCR assay was used to detect EV genome in RNA extracted from the specimens and controls.Results: The assay detected EV RNA in a 10−5 dilution of infected mouse tissue. EV RNA was not detected in the ALS specimens or in specimens from control cases, despite the presence of amplifiable RNA as assessed by amplification with control primers, whereas all of the positive control specimens yielded the expected PV amplification product.Conclusion: The reported association between EV infection and ALS was not confirmed by testing this set of specimens with these sensitive methods.
- Published
- 2004
- Full Text
- View/download PDF
28. Superoxide Dismutase Concentration and Activity in Familial Amyotrophic Lateral Sclerosis
- Author
-
Robert H. Brown, HR Horvitz, Diane McKenna-Yasek, Bowling Ac, Barkowski Ee, M F Beal, and Peter C. Sapp
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pathology ,Erythrocytes ,SOD1 ,medicine.disease_cause ,Biochemistry ,Pathogenesis ,Superoxide dismutase ,Cellular and Molecular Neuroscience ,Internal medicine ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Aged ,biology ,Superoxide Dismutase ,Chemistry ,Amyotrophic Lateral Sclerosis ,Neurodegeneration ,Middle Aged ,medicine.disease ,Endocrinology ,Mutation ,biology.protein ,Regression Analysis ,Female ,Dismutase ,Specific activity ,Oxidative stress - Abstract
Some cases of autosomal-dominant familial amyotrophic lateral sclerosis (FALS) have been associated with mutations in SOD1, the gene that encodes Cu/Zn superoxide dismutase (Cu/Zn SOD). We determined the concentrations (microgram of Cu/Zn SOD/mg of total protein), specific activities (U/microgram of total protein), and apparent turnover numbers (U/mumol of Cu/Zn SOD) of Cu/Zn SOD in erythrocyte lysates from patients with known SOD1 mutations. We also measured the concentrations and activities of Cu/Zn SOD in FALS patients with no identifiable SOD1 mutations, sporadic ALS (SALS) patients, and patients with other neurologic disorders. The concentration and specific activity of Cu/Zn SOD were decreased in all patients with SOD1 mutations, with mean reductions of 51 and 46%, respectively, relative to controls. In contrast, the apparent turnover number of the enzyme was not altered in these patients. For the six mutations studied, there was no correlation between enzyme concentration or specific activity and disease severity, expressed as either duration of disease or age of onset. No significant alterations in the concentration, specific activity, or apparent turnover number of Cu/Zn SOD were detected in the FALS patients with no identifiable SOD1 mutations, SALS patients, or patients with other neurologic disorders. That Cu/Zn SOD concentration and specific activity are equivalently reduced in erythrocytes from patients with SOD1 mutations suggests that mutant Cu/Zn SOD is unstable in these cells. That concentration and specific activity do not correlate with disease severity suggests that an altered, novel function of the enzyme, rather than reduction of its dismutase activity, may be responsible for the pathogenesis of FALS.
- Published
- 2002
- Full Text
- View/download PDF
29. Identifying diagnostic DNA methylation profiles for facioscapulohumeral muscular dystrophy in blood and saliva using bisulfite sequencing
- Author
-
Johnny Salameh, Takako I. Jones, Chi Yan, Peter L. Jones, Peter C. Sapp, Peter B. Kang, Diane McKenna-Yasek, Colin Quinn, and Oliver D. King
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,DUX4 ,Bisulfite sequencing ,Biology ,FSHD2 ,Facioscapulohumeral muscular dystrophy ,Genetics ,medicine ,Epigenetics ,Molecular Biology ,Genetics (clinical) ,Southern blot ,DNA methylation ,D4Z4 ,Disease diagnostics ,Methodology ,medicine.disease ,Subtelomere ,Molecular biology ,3. Good health ,FSHD1 ,genomic DNA ,Developmental Biology ,DNA hypomethylation - Abstract
Background Facioscapulohumeral muscular dystrophy (FSHD) is linked to chromatin relaxation due to epigenetic changes at the 4q35 D4Z4 macrosatellite array. Molecular diagnostic criteria for FSHD are complex and involve analysis of high molecular weight (HMW) genomic DNA isolated from lymphocytes, followed by multiple restriction digestions, pulse-field gel electrophoresis (PFGE), and Southern blotting. A subject is genetically diagnosed as FSHD1 if one of the 4q alleles shows a contraction in the D4Z4 array to below 11 repeats, while maintaining at least 1 repeat, and the contraction is in cis with a disease-permissive A-type subtelomere. FSHD2 is contraction-independent and cannot be diagnosed or excluded by this common genetic diagnostic procedure. However, FSHD1 and FSHD2 are linked by epigenetic deregulation, assayed as DNA hypomethylation, of the D4Z4 array on FSHD-permissive alleles. We have developed a PCR-based assay that identifies the epigenetic signature for both types of FSHD, distinguishing FSHD1 from FSHD2, and can be performed on genomic DNA isolated from blood, saliva, or cultured cells. Results Samples were obtained from healthy controls or patients clinically diagnosed with FSHD, and include both FSHD1 and FSHD2. The genomic DNAs were subjected to bisulfite sequencing analysis for the distal 4q D4Z4 repeat with an A-type subtelomere and the DUX4 5’ promoter region. We compared genomic DNA isolated from saliva and blood from the same individuals and found similar epigenetic signatures. DNA hypomethylation was restricted to the contracted 4qA chromosome in FSHD1 patients while healthy control subjects were hypermethylated. Candidates for FSHD2 showed extreme DNA hypomethylation on the 4qA DUX4 gene body as well as all analyzed DUX4 5’ sequences. Importantly, our assay does not amplify the D4Z4 arrays with non-permissive B-type subtelomeres and accurately excludes the arrays with non-permissive A-type subtelomeres. Conclusions We have developed an assay to identify changes in DNA methylation on the pathogenic distal 4q D4Z4 repeat. We show that the DNA methylation profile of saliva reflects FSHD status. This assay can distinguish FSHD from healthy controls, differentiate FSHD1 from FSHD2, does not require HMW genomic DNA or PFGE, and can be performed on either cultured cells, tissue, blood, or saliva samples. Electronic supplementary material The online version of this article (doi:10.1186/1868-7083-6-23) contains supplementary material, which is available to authorized users.
- Published
- 2014
30. A novel dysferlin mutant pseudoexon bypassed with antisense oligonucleotides
- Author
-
Robert H. Brown, Diane McKenna-Yasek, Peter C. Sapp, Özgün Uyan, Babi Ramesh Reddy Nallamilli, Janice A. Dominov, and Madhuri Hegde
- Subjects
Genetics ,0303 health sciences ,Messenger RNA ,Mutation ,Dysferlinopathy ,biology ,General Neuroscience ,Mutant ,Context (language use) ,medicine.disease_cause ,medicine.disease ,3. Good health ,Dysferlin ,03 medical and health sciences ,Exon ,0302 clinical medicine ,RNA splicing ,biology.protein ,medicine ,Neurology (clinical) ,030217 neurology & neurosurgery ,Research Articles ,030304 developmental biology - Abstract
Objective Mutations in dysferlin (DYSF), a Ca2+-sensitive ferlin family protein important for membrane repair, vesicle trafficking, and T-tubule function, cause Miyoshi myopathy, limb-girdle muscular dystrophy type 2B, and distal myopathy. More than 330 pathogenic DYSF mutations have been identified within exons or near exon–intron junctions. In ~17% of patients who lack normal DYSF, only a single disease-causing mutation has been identified. We studied one family with one known mutant allele to identify both the second underlying genetic defect and potential therapeutic approaches. Methods We sequenced the full DYSF cDNA and investigated antisense oligonucleotides (AONs) as a tool to modify splicing of the mRNA transcripts in order to process out mutant sequences. Results We identified a novel pseudoexon between exons 44 and 45, (pseudoexon 44.1, PE44.1), which inserts an additional 177 nucleotides into the mRNA and 59 amino acids within the conserved C2F domain of the DYSF protein. Two unrelated dysferlinopathy patients were also found to carry this mutation. Using AONs targeting PE44.1, we blocked the abnormal splicing event, yielding normal, full-length DYSF mRNA, and increased DYSF protein expression. Interpretation This is the first report of a deep intronic mutation in DYSF that alters mRNA splicing to include a mutant peptide fragment within a key DYSF domain. We report that AON-mediated exon-skipping restores production of normal, full-length DYSF in patients' cells in vitro, offering hope that this approach will be therapeutic in this genetic context, and providing a foundation for AON therapeutics targeting other pathogenic DYSF alleles.
- Published
- 2014
31. Partial loss of TDP-43 function causes phenotypes of amyotrophic lateral sclerosis
- Author
-
Leonardo Aliaga, Chunxing Yang, Linghua Qiu, Bin Yang, Weijia Tan, Tao Qiao, Hongyan Wang, Melissa J. Moore, Diane McKenna-Yasek, Thomas W. Smith, Lingtao Peng, Johnny Salameh, Huaibin Cai, Robert H. Brown, and Zuoshang Xu
- Subjects
Genetically modified mouse ,Central Nervous System ,Transgene ,Central nervous system ,Biology ,Pathogenesis ,Animals, Genetically Modified ,Mice ,mental disorders ,Paralysis ,medicine ,Animals ,Amyotrophic lateral sclerosis ,Gene knockdown ,Multidisciplinary ,Amyotrophic Lateral Sclerosis ,nutritional and metabolic diseases ,medicine.disease ,Spinal cord ,nervous system diseases ,DNA-Binding Proteins ,Disease Models, Animal ,medicine.anatomical_structure ,Phenotype ,PNAS Plus ,medicine.symptom ,Neuroscience - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease that causes motor neuron degeneration, progressive motor dysfunction, paralysis, and death. Although multiple causes have been identified for this disease, >95% of ALS cases show aggregation of transactive response DNA binding protein (TDP-43) accompanied by its nuclear depletion. Therefore, the TDP-43 pathology may be a converging point in the pathogenesis that originates from various initial triggers. The aggregation is thought to result from TDP-43 misfolding, which could generate cellular toxicity. However, the aggregation as well as the nuclear depletion could also lead to a partial loss of TDP-43 function or TDP-43 dysfunction. To investigate the impact of TDP-43 dysfunction, we generated a transgenic mouse model for a partial loss of TDP-43 function using transgenic RNAi. These mice show ubiquitous transgene expression and TDP-43 knockdown in both the periphery and the central nervous system (CNS). Strikingly, these mice develop progressive neurodegeneration prominently in cortical layer V and spinal ventral horn, motor dysfunction, paralysis, and death. Furthermore, examination of splicing patterns of TDP-43 target genes in human ALS revealed changes consistent with TDP-43 dysfunction. These results suggest that the CNS, particularly motor neurons, possess a heightened vulnerability to TDP-43 dysfunction. Additionally, because TDP-43 knockdown predominantly occur in astrocytes in the spinal cord of these mice, our results suggest that TDP-43 dysfunction in astrocytes is an important driver for motor neuron degeneration and clinical phenotypes of ALS.
- Published
- 2014
32. Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy
- Author
-
Diane McKenna-Yasek, Robert H. Brown, J. Andoni Urtizberea, Edward J. Culper, K. Bossie, Fayçal Hentati, Khemissa Bejaoui, Corrado Angelini, Erwin Schurr, Jing Liu, Chenyan Wu, Masashi Aoki, Joshua Oeltjen, Isabel Illa, Mongi Ben Hamida, Betsy A. Hosler, Anthony A. Amato, Saeed Bohlega, Carmen Serrano, Kiichi Arahata, Michel Fardeau, and Pieter J. de Jong
- Subjects
Adult ,Genetic Markers ,Male ,Dysferlinopathy ,Molecular Sequence Data ,Gene Expression ,Muscle Proteins ,Muscular Dystrophies ,Dysferlin ,Gene mapping ,Genetics ,medicine ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Muscle, Skeletal ,Myopathy ,Polymorphism, Single-Stranded Conformational ,Polymorphism, Genetic ,biology ,Membrane Proteins ,Dystrophy ,Blotting, Northern ,medicine.disease ,Mutation ,biology.protein ,Female ,medicine.symptom ,Distal muscular dystrophy ,Limb-girdle muscular dystrophy ,Sarcoglycanopathies - Abstract
Miyoshi myopathy (MM) is an adult onset, recessive inherited distal muscular dystrophy that we have mapped to human chromosome 2p13. We recently constructed a 3-Mb P1-derived artificial chromosome (PAC) contig spanning the MM candidate region. This clarified the order of genetic markers across the MM locus, provided five new polymorphic markers within it and narrowed the locus to approximately 2 Mb. Five skeletal muscle expressed sequence tags (ESTs) map in this region. We report that one of these is located in a novel, full-length 6.9-kb muscle cDNA, and we designate the corresponding protein 'dysferlin'. We describe nine mutations in the dysferlin gene in nine families; five are predicted to prevent dysferlin expression. Identical mutations in the dysferlin gene can produce more than one myopathy phenotype (MM, limb girdle dystrophy, distal myopathy with anterior tibial onset).
- Published
- 1998
- Full Text
- View/download PDF
33. Identification of two mutations and a polymorphism in the chloride channel CLCN-1 in patients with Becker's generalized myotonia
- Author
-
Robert H. Brown, Jesus Esteban, Diane McKenna-Yasek, and Ann M. Neumeyer
- Subjects
Male ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Myotonia Congenita ,DNA Mutational Analysis ,Muscle disorder ,Muscle hypertrophy ,Cellular and Molecular Neuroscience ,Chloride Channels ,Genetics ,medicine ,Humans ,Point Mutation ,Genetics (clinical) ,Family Health ,CLCN1 ,Polymorphism, Genetic ,biology ,Myotonia congenita ,Point mutation ,Skeletal muscle ,DNA ,Myotonia ,medicine.disease ,Pedigree ,Mutagenesis, Insertional ,medicine.anatomical_structure ,Amino Acid Substitution ,Mutation ,biology.protein ,Chloride channel ,Female - Abstract
Myotonia congenita is an inherited muscle disorder characterized by muscle stiffness and hypertrophy. Its clinical phenotype depends, in part, on whether it is inherited as a dominant or recessive trait, respectively designated Thomsen's disease or Becker's generalized myotonia (BGM). In either case, it is associated with abnormalities in the muscle currents that are linked to the gene (CLCN-1) on human chromosome 7q35 encoding the skeletal muscle chloride channel. Single-strand conformation polymorphism analysis was used to screen two families with the BGM for mutations in the CLCN-1 gene. Two new mutations were found (G 201ins and A317Q). The latter mutation has been previously described in Thomsen's disease.
- Published
- 1998
- Full Text
- View/download PDF
34. Genetic fine mapping of the Miyoshi myopathy locus and exclusion of eight candidate genes
- Author
-
Diane McKenna-Yasek, Robert H. Brown, D. M. Gilligan, D. Le Paslier, K. Bossie, Khemissa Bejaoui, and Jing Liu
- Subjects
Male ,Yeast artificial chromosome ,Candidate gene ,Genotype ,Genetic Linkage ,Locus (genetics) ,Biology ,Muscular Dystrophies ,Contig Mapping ,Cellular and Molecular Neuroscience ,Genetic linkage ,Genetics ,Humans ,Genetic Predisposition to Disease ,Chromosomes, Artificial, Yeast ,Genetics (clinical) ,Family Health ,Polymorphism, Genetic ,Contig ,Haplotype ,Chromosome Mapping ,DNA ,Pedigree ,Haplotypes ,Genetic marker ,Microsatellite ,Calmodulin-Binding Proteins ,Female ,Microsatellite Repeats - Abstract
Miyoshi myopathy (MM) is an early adult-onset, autosomal recessive disorder characterized by weakness and muscular atrophy starting in the distal muscles. The disease locus has been previously mapped by linkage analysis to chromosome 2p using the microsatellite marker D2S291. Initial haplotype analysis of markers in families from three different origins (North American, Japanese, and Tunisian) suggested that the MM gene is located in a 4-cM region flanked by markers D2S292 on the telomeric side and D2S286 on the centromeric side. To delineate critical recombination events revealing a more refined localization of the MM gene, we have determined the pattern of segregation of 12 marker loci in two consanguineous families of Tunisian origin. In this study we have: (1) detected recombination events with the disease locus in one family, placing the MM gene most likely between markers D2S443 (CHLC.GGAA4D07.1876) and D2S2109; (2) generated a yeast artificial chromosome contig that spans approximately 3.8 megabases and extends from marker D2S358 to marker D2S286; (3) physically mapped 21 polymorphic markers, 5 genes, 3 STSs, and 1 EST within this contig; (4) detected and mapped a new polymorphism within this interval, allowing us to further reduce the MM locus to a 360-kilobase segment; (5) mapped the gene for the cytoskeletal protein beta-adducin within the MM candidate region, failing to find a consistent pattern of mutation of this gene in our MM patients; (6) excluded seven other candidate myopathy genes from the Miyoshi locus.
- Published
- 1998
- Full Text
- View/download PDF
35. Exome-wide rare variant analysis identifies TUBA4A mutations associated with familial ALS
- Author
-
Bradley N. Smith, Nicola Ticozzi, Claudia Fallini, Athina Soragia Gkazi, Simon Topp, Kevin P. Kenna, Emma L. Scotter, Jason Kost, Pamela Keagle, Jack W. Miller, Daniela Calini, Caroline Vance, Eric W. Danielson, Claire Troakes, Cinzia Tiloca, Safa Al-Sarraj, Elizabeth A. Lewis, Andrew King, Claudia Colombrita, Viviana Pensato, Barbara Castellotti, Jacqueline de Belleroche, Frank Baas, Anneloor LMA ten Asbroek, Peter C. Sapp, Diane McKenna-Yasek, Russell L. McLaughlin, Meraida Polak, Seneshaw Asress, Jesús Esteban-Pérez, José Luis Muñoz-Blanco, Michael Simpson, Wouter van Rheenen, Frank P. Diekstra, Giuseppe Lauria, Stefano Duga, Stefania Corti, Cristina Cereda, Lucia Corrado, Gianni Sorarù, Karen E. Morrison, Kelly L. Williams, Garth A. Nicholson, Ian P. Blair, Patrick A. Dion, Claire S. Leblond, Guy A. Rouleau, Orla Hardiman, Jan H. Veldink, Leonard H. van den Berg, Ammar Al-Chalabi, Hardev Pall, Pamela J. Shaw, Martin R. Turner, Kevin Talbot, Franco Taroni, Alberto García-Redondo, Zheyang Wu, Jonathan D. Glass, Cinzia Gellera, Antonia Ratti, Robert H. Brown, Vincenzo Silani, Christopher E. Shaw, John E. Landers, Sandra D’Alfonso, Letizia Mazzini, Giacomo P. Comi, Roberto Del Bo, Mauro Ceroni, Stella Gagliardi, Giorgia Querin, Cinzia Bertolin, ANS - Amsterdam Neuroscience, Genome Analysis, and Human Genetics
- Subjects
Neurons ,Genetics ,Mutation ,Sequence analysis ,General Neuroscience ,Neuroscience(all) ,Amyotrophic Lateral Sclerosis ,Brain ,Sequence Analysis, DNA ,Disease ,Biology ,medicine.disease ,medicine.disease_cause ,Article ,Tubulin ,medicine ,Missense mutation ,Humans ,Exome ,Genetic Predisposition to Disease ,Amyotrophic lateral sclerosis ,Gene ,Exome sequencing - Abstract
SummaryExome sequencing is an effective strategy for identifying human disease genes. However, this methodology is difficult in late-onset diseases where limited availability of DNA from informative family members prohibits comprehensive segregation analysis. To overcome this limitation, we performed an exome-wide rare variant burden analysis of 363 index cases with familial ALS (FALS). The results revealed an excess of patient variants within TUBA4A, the gene encoding the Tubulin, Alpha 4A protein. Analysis of a further 272 FALS cases and 5,510 internal controls confirmed the overrepresentation as statistically significant and replicable. Functional analyses revealed that TUBA4A mutants destabilize the microtubule network, diminishing its repolymerization capability. These results further emphasize the role of cytoskeletal defects in ALS and demonstrate the power of gene-based rare variant analyses in situations where causal genes cannot be identified through traditional segregation analysis.Video Abstract
- Published
- 2014
36. Three novel mutations and two variants in the gene for Cu/Zn superoxide dismutase in familial amyotrophic lateral sclerosis
- Author
-
Garth A. Nicholson, Betsy A. Hosler, Diane McKenna-Yasek, Jesus Esteban, Steve D. Wilton, A.K. Cherryson, J. de Belleroche, Peter C. Sapp, Lawrence J. Hayward, H. R. Horvitz, J.E. Dench, William W. Chin, Nigel G. Laing, L. Yeung, Robert H. Brown, and Richard W. Orrell
- Subjects
SOD1 ,Biology ,medicine.disease_cause ,Exon ,Valine ,medicine ,Humans ,Point Mutation ,Missense mutation ,Amyotrophic lateral sclerosis ,Gene ,Polymorphism, Single-Stranded Conformational ,Genetics (clinical) ,Family Health ,Genetics ,Mutation ,Polymorphism, Genetic ,Superoxide Dismutase ,Point mutation ,Amyotrophic Lateral Sclerosis ,nutritional and metabolic diseases ,Sequence Analysis, DNA ,medicine.disease ,Molecular biology ,Neurology ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) - Abstract
Autosomal dominant inheritance is exhibited by about 10% of cases of amyotrophic lateral sclerosis (ALS), a paralytic disorder characterized by the death of motor neurons in the brain and spinal cord. A subgroup of these familial cases are linked to mutations in the gene which codes for Cu/Zn superoxide dismutase (SOD1). We report three additional mutations occurring in the SOD1 gene in ALS patients and two single base pair variant changes. The single base pair change in an ALS family causes a glycine 93 to valine substitution, which is the fifth distinct amino acid change reported for the glycine 93 residue. One missense mutation in exon 5 would substitute neutral valine for the negatively-charged aspartate 124 (aspartate 124 to valine). An individual with an apparently sporadic case of ALS carries a three base pair deletion in exon 5 of the SOD1 gene. These three mutations bring to 38 the total number of distinct SOD1 mutations associated with familial ALS.
- Published
- 1996
- Full Text
- View/download PDF
37. Identification of three novel mutations in the gene for CuZn superoxide dismutase in patients with familial amyotrophic lateral sclerosis
- Author
-
Robert H. Brown, Jeremiah P. O'Regan, Peter C. Sapp, Daniel R. Rosen, Jesus Esteban, Diane McKenna-Yasek, H. Robert Horvitz, and Betsy A. Hosler
- Subjects
Adult ,RNA Splicing ,Molecular Sequence Data ,SOD1 ,Biology ,Polymerase Chain Reaction ,Serine ,Open Reading Frames ,Exon ,medicine ,Humans ,Missense mutation ,Age of Onset ,Amyotrophic lateral sclerosis ,Gene ,Polymorphism, Single-Stranded Conformational ,Genetics (clinical) ,Aged ,Genes, Dominant ,Genetics ,Base Sequence ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,Intron ,Exons ,Middle Aged ,medicine.disease ,Molecular biology ,Open reading frame ,Neurology ,Mutation ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) - Abstract
About 10% of cases of amyotrophic lateral sclerosis (ALS), a paralytic disorder characterized by death of motor neurons in the brain and spinal cord, exhibit autosomal dominant inheritance. A subgroup of these familial cases are caused by mutations in the gene encoding Cu Zn superoxide dismutase (SOD1). We report here three additional mutations occurring in the SOD1 gene in three families with ALS. Two of these changes are missense mutations in exon 5 of the SOD1 gene, resulting in leucine 144 to serine and alanine 145 to threonine substitutions. The third, a single base pair change in intron 4 immediately upstream of exon 5, results in an alternatively spliced mRNA. The alternate transcript conserves the open reading frame of exon 5, producing an SOD1 protein with three amino acids inserted between exons 4 and 5 (following residue 118). These three mutations bring to 29 the total number of distinct SOD1 mutations associated with familial ALS.
- Published
- 1995
- Full Text
- View/download PDF
38. Novel mutation in VCP gene causes atypical amyotrophic lateral sclerosis
- Author
-
Menachem Sadeh, Robert H. Brown, Elizabeth T. Cirulli, Peter C. Sapp, P. Nisipeanu, Barry W. Festoff, Paloma Gonzalez-Perez, David Goldstein, Andrew Fox, R.L. Carasso, Ron Dabby, Sergiu C. Blumen, Vivian E. Drory, and Diane McKenna-Yasek
- Subjects
Adult ,Male ,Genetic Linkage ,Valosin-containing protein ,Single-nucleotide polymorphism ,Cell Cycle Proteins ,Gene mutation ,medicine.disease_cause ,Article ,Genetic linkage ,Valosin Containing Protein ,medicine ,Humans ,Point Mutation ,Amyotrophic lateral sclerosis ,Exome sequencing ,Genetics ,Adenosine Triphosphatases ,Family Health ,Mutation ,biology ,Point mutation ,Amyotrophic Lateral Sclerosis ,Middle Aged ,medicine.disease ,Pedigree ,biology.protein ,Female ,Neurology (clinical) - Abstract
Objective: To identify the genetic variant that causes autosomal dominantly inherited motor neuron disease in a 4-generation Israeli-Arab family using genetic linkage and whole exome sequencing. Methods: Genetic linkage analysis was performed in this family using Illumina single nucleotide polymorphism chips. Whole exome sequencing was the nu ndertaken on DNA samples from 2a ffected family members using an Illumina 2000 HiSeq platform in pursuit of potentially pathogenic genetic variants that comigrate with the disease in this pedigree. Variants meeting these criteria were then screened in all affected individuals. Results: A novel mutation (p.R191G) in the valosin-containing protein (VCP) gene was identified in the index family. Direct sequencing of the VCP gene in a panel of DNA from 274 unrelated individuals with familial amyotrophic lateral sclerosis (FALS) revealed 5 additional mutations. Among them, 2 were previously identified in pedigrees with a constellation of inclusion body myopathy with Paget disease of the bone and frontotemporal dementia (IBMPFD) and in FALS, and 2 other mutations (p.R159C and p.R155C) in IBMPFD alone. We did not detect VCP gene mutations in DNA from 178 cases of sporadic amyotrophic lateral sclerosis. Conclusions: We report a novel VCP mutation identified in an amyotrophic lateral sclerosis family (p.R191G) with atypical clinical features. In our experience, VCP mutations arise in approximately 1.5% of FALS cases. Our study supports the view that motor neuron disease is part of the clinical spectrum of VCP-associated disease. Neurology � 2012;79:2201–2208
- Published
- 2012
39. Association of UBQLN1 mutation with Brown-Vialetto-Van Laere syndrome but not typical ALS
- Author
-
Fen-Biao Gao, Peter C. Sapp, Robert H. Brown, Lars Bertram, Diane McKenna-Yasek, Paloma Gonzalez-Perez, Yubing Lu, Ru-Ju Chian, and Rudolph E. Tanzi
- Subjects
Male ,TDP-43 ,Hearing Loss, Sensorineural ,Blotting, Western ,Bulbar Palsy, Progressive ,DNA Mutational Analysis ,Autophagy-Related Proteins ,Cell Cycle Proteins ,Gene mutation ,medicine.disease_cause ,Transfection ,Polymorphism, Single Nucleotide ,Article ,lcsh:RC321-571 ,UBQLN2 ,Brown–Vialetto–Van Laere syndrome ,medicine ,Missense mutation ,Humans ,Immunoprecipitation ,Ubiquilins ,Amyotrophic lateral sclerosis ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Gene ,Adaptor Proteins, Signal Transducing ,Genetics ,Mutation ,biology ,Neurodegeneration ,Amyotrophic Lateral Sclerosis ,Drosophila motor neuron disease ,medicine.disease ,Molecular biology ,Immunohistochemistry ,Neurology ,biology.protein ,Female ,Carrier Proteins - Abstract
Genetic variants in UBQLN1 gene have been linked to neurodegeneration and mutations in UBQLN2 have recently been identified as a rare cause of amyotrophic lateral sclerosis (ALS). Objective To test if genetic variants in UBQLN1 are involved in ALS. Methods 102 and 94 unrelated patients with familial and sporadic forms of ALS were screened for UBQLN1 gene mutations. Single nucleotide variants were further screened in a larger set of sporadic ALS (SALS) patients and unrelated control subjects using high-throughput Taqman genotyping; variants were further assessed for novelty using the 1000Genomes and NHLBI databases. In vitro studies tested the effect of UBQLN1 variants on the ubiquitin–proteasome system (UPS). Results Only two UBQLN1 coding variants were detected in the familial and sporadic ALS DNA set; one, the missense mutation p.E54D, was identified in a single patient with atypical motor neuron disease consistent with Brown–Vialetto–Van Laere syndrome (BVVLS), for whom c20orf54 mutations had been excluded. Functional studies revealed that UBQLN1 E54D protein forms cytosolic aggregates that contain mislocalized TDP-43 and impairs degradation of ubiquitinated proteins through the proteasome. Conclusions Genetic variants in UBQLN1 are not commonly associated with ALS. A novel UBQLN1 mutation (E45D) detected in a patient with BVVLS altered nuclear TDP-43 localization in vitro , suggesting that UPS dysfunction may also underlie the pathogenesis of this condition.
- Published
- 2012
40. Mutations in the profilin 1 gene cause familial amyotrophic lateral sclerosis
- Author
-
Desiree M. Baron, Max Koppers, Gary J. Bassell, Andrew Fox, Elizabeth T. Cirulli, Chi Hong Wu, Dev Mangroo, Zuoshang Xu, Jill A. Zitzewitz, Cinzia Gellera, David Goldstein, John Landers, Vivian E. Drory, Diane McKenna-Yasek, François Salachas, Wilfried Rossoll, Paloma Gonzalez-Perez, Robert H. Brown, Katarzyna Piotrowska, Leonard H. van den Berg, Ashley Lyn Leclerc, Patrick Lowe, Franco Taroni, Pamela Keagle, Jenni Adams, Peter C. Sapp, Shawn C. Chafe, Claudia Fallini, Nicola Ticozzi, Antonia Ratti, Jonathan D. Glass, Vincenzo Silani, Gabriele Siciliano, Jason E. Kost, Melissa J. Moore, Cinzia Tiloca, Daryl A. Bosco, and Vincent Meininger
- Subjects
Male ,Models, Molecular ,Protein Conformation ,Growth Cones ,Molecular Sequence Data ,Mutant ,medicine.disease_cause ,White People ,Article ,Mice ,Profilins ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Exome ,Genetic Predisposition to Disease ,Amino Acid Sequence ,Amyotrophic lateral sclerosis ,Gene ,Cells, Cultured ,Exome sequencing ,Actin ,030304 developmental biology ,Motor Neurons ,Genetics ,0303 health sciences ,Mutation ,Multidisciplinary ,biology ,Amyotrophic Lateral Sclerosis ,Ubiquitination ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Actins ,Axons ,Pedigree ,Profilin ,Jews ,biology.protein ,Female ,Mutant Proteins ,030217 neurology & neurosurgery - Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset neurodegenerative disorder resulting from motor neuron death. Approximately 10% of cases are familial (FALS), typically with a dominant inheritance mode. Despite numerous advances in recent years1-9, nearly 50% of FALS cases have unknown genetic etiology. Here we show that mutations within the profilin 1 (PFN1) gene can cause FALS. PFN1 is critical for monomeric (G)-actin conversion to filamentous (F)-actin. Exome sequencing of two large ALS families revealed different mutations within the PFN1 gene. Additional sequence analysis identified 4 mutations in 7 out of 274 FALS cases. Cells expressing PFN1 mutants contain ubiquitinated, insoluble aggregates that in many cases contain the ALS-associated protein TDP-43. PFN1 mutants also display decreased bound actin levels and can inhibit axon outgrowth. Furthermore, primary motor neurons expressing mutant PFN1 display smaller growth cones with a reduced F-/G-actin ratio. These observations further document that cytoskeletal pathway alterations contribute to ALS pathogenesis.
- Published
- 2012
41. Genetic linkage analysis of familial amyotrophic lateral sclerosis using human chromosome 21 microsatellite DNA markers
- Author
-
Daniel R. Rosen, Jonathan L. Haines, Diane McKenna-Yasek, Karen Schlumpf, Peter C. Sapp, James F. Gusella, Jeremiah P. O'Regan, Robert H. Brown, and HR Horvitz
- Subjects
Genetic Markers ,Male ,Genetic Linkage ,DNA Mutational Analysis ,Molecular Sequence Data ,Chromosomes, Human, Pair 20 ,DNA, Satellite ,Gene mutation ,Biology ,Gene mapping ,Genetic linkage ,Humans ,Point Mutation ,Genetics (clinical) ,DNA Primers ,Repetitive Sequences, Nucleic Acid ,Genetics ,Base Sequence ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,Haplotype ,Chromosome Mapping ,Molecular biology ,Pedigree ,Haplotypes ,Genetic marker ,Microsatellite ,Female ,Lod Score ,Restriction fragment length polymorphism ,Chromosome 21 ,Dinucleoside Phosphates ,Polymorphism, Restriction Fragment Length - Abstract
Amyotrophic lateral sclerosis (ALS: Lou Gehrig's Disease) is a lethal neurodegenerative disease of upper and lower motorneurons in the brain and spinal cord. We previously reported linkage of a gene for familial ALS (FALS) to human chromosome 21 using 4 restriction fragment length polymorphism DNA markers [Siddique et al.: N Engl J Med 324:1381-1384, 1991] and identified disease-associated mutations in the superoxide dismutase (SOD)-1 gene in some ALS families [Rosen et al.: Nature 362:59-62, 1993]. We report here the genetic linkage data that led us to examine the SOD-1 gene for mutations. We also report a new microsatellite DNA marker for D21S63, derived from the cosmid PW517 [VanKeuren et al.: Am J Hum Genet 38:793-804, 1986]. Ten microsatellite DNA markers, including the new marker D21S63, were used to reinvestigate linkage of FALS to chromosome 21. Genetic linkage analysis performed with 13 ALS families for these 10 DNA markers confirmed the presence of a FALS gene on chromosome 21. The highest total 2-point LOD score for all families was 4.33, obtained at a distance of 10 cM from the marker D21S223. For 5 ALS families linked to chromosome 21, a peak 2-point LOD score of 5.94 was obtained at the DNA marker D21S223. A multipoint score of 6.50 was obtained with the markers D21S213, D21S223, D21S167, and FALS for 5 chromosome 21-linked ALS families. The haplotypes of these families for the 10 DNA markers revealed recombination events that further refined the location of the FALS gene to a segment of approximately 5 megabases (Mb) between D21S213 and D21S219.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1994
- Full Text
- View/download PDF
42. A frequent ala 4 to val superoxide dismutase-1 mutation is associated with a rapidly progressive familial amyotrophic lateral sclerosis
- Author
-
Zohra Rahmani, D. Patterson, Robert H. Brown, Robert J. Ferrante, Eva Mezey, Bowling Ac, Diane McKenna-Yasek, Daniel R. Rosen, Jerimiah O'Regan, Peter C. Sapp, M. Flint Beal, H. Robert Horvitz, Ted B. Usdin, Neil W. Kowall, and Michael J. Brownstein
- Subjects
medicine.medical_specialty ,animal diseases ,Molecular Sequence Data ,SOD1 ,Biology ,medicine.disease_cause ,Polymerase Chain Reaction ,Superoxide dismutase ,Exon ,Degenerative disease ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Point Mutation ,Amino Acid Sequence ,Age of Onset ,Amyotrophic lateral sclerosis ,Molecular Biology ,In Situ Hybridization ,Genetics (clinical) ,DNA Primers ,Mutation ,Alanine ,Sequence Homology, Amino Acid ,Superoxide Dismutase ,Point mutation ,Amyotrophic Lateral Sclerosis ,nutritional and metabolic diseases ,Valine ,Exons ,General Medicine ,Middle Aged ,Motor neuron ,medicine.disease ,Survival Analysis ,nervous system diseases ,Isoenzymes ,medicine.anatomical_structure ,Endocrinology ,nervous system ,biology.protein - Abstract
Familial amyotrophic lateral sclerosis (FALS), a degenerative disorder of motor neurons, is associated with mutations in the Cu/Zn superoxide dismutase gene SOD1 in some affected families. We confirm a recently reported ala4-->val mutation in exon 1 of the SOD1 gene and report that this mutation is both the most commonly detected of all SOD1 mutations and among the most clinically severe. By comparison with our other FALS families, the exon 1 mutation is associated with reduced survival time after onset: 1.2 years, as compared to 2.5 years for all other FALS patients. We also demonstrate that SOD1 is prominently expressed in normal motor neurons and that neural expression of SOD1 is not prevented by this exon 1 mutation. Assays of SOD1 enzymatic activity in extracts from red blood cells, lymphoblastoid cells, and brain tissues revealed an approximately 50% reduction in activity of cytosolic SOD1 in patients with this mutation compared to normal individuals. By contrast, patients with sporadic ALS had normal levels of SOD1 enzymatic activity. Why this SOD1 mutation causes motor neuron death in FALS remains to be established. While it may be that FALS is a consequence of loss of SOD1 function, it is also possible that motor neuron death in this dominantly inherited disease occurs because the mutations confer an additional, cytotoxic function on the SOD1 protein.
- Published
- 1994
- Full Text
- View/download PDF
43. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis
- Author
-
Daniel R. Rosen, Teepu Siddique, David Patterson, Denise A. Figlewicz, Peter Sapp, Afif Hentati, Deirdre Donaldson, Jun Goto, Jeremiah P. O'Regan, Han-Xiang Deng, Zohra Rahmani, Aldis Krizus, Diane McKenna-Yasek, Annarueber Cayabyab, Sandra M. Gaston, Ralph Berger, Rudolph E. Tanzi, John J. Halperin, Brian Herzfeldt, Raymond Van den Bergh, Wu-Yen Hung, Thomas Bird, Gang Deng, Donald W. Mulder, Celestine Smyth, Nigel G. Laing, Edwin Soriano, Margaret A. Pericak–Vance, Jonathan Haines, Guy A. Rouleau, James S. Gusella, H. Robert Horvitz, and Robert H. Brown
- Subjects
Juvenile amyotrophic lateral sclerosis ,Molecular Sequence Data ,SOD1 ,Polymerase Chain Reaction ,TARDBP ,UBQLN2 ,C9orf72 ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Amyotrophic lateral sclerosis ,Codon ,Repetitive Sequences, Nucleic Acid ,Genetics ,Polymorphism, Genetic ,Multidisciplinary ,Base Sequence ,Sequence Homology, Amino Acid ,biology ,Genome, Human ,Superoxide Dismutase ,Amyotrophic Lateral Sclerosis ,Autosomal dominant trait ,DNA ,Exons ,medicine.disease ,Isoenzymes ,Copper chaperone for superoxide dismutase ,Oligodeoxyribonucleotides ,Mutation ,biology.protein ,Nucleic Acid Conformation ,Lod Score ,biology.gene - Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerative disorder of motor neurons in the cortex, brainstem and spinal cord. Its cause is unknown and it is uniformly fatal, typically within five years. About 10% of cases are inherited as an autosomal dominant trait, with high penetrance after the sixth decade. In most instances, sporadic and autosomal dominant familial ALS (FALS) are clinically similar. We have previously shown that in some but not all FALS pedigrees the disease is linked to a genetic defect on chromosome 21q (refs 8, 9). Here we report tight genetic linkage between FALS and a gene that encodes a cytosolic, Cu/Zn-binding superoxide dismutase (SOD1), a homodimeric metalloenzyme that catalyzes the dismutation of the toxic superoxide anion O2.- to O2 and H2O2 (ref. 10). Given this linkage and the potential role of free radical toxicity in other neurodenegerative disorders, we investigated SOD1 as a candidate gene in FALS. We identified 11 different SOD1 missense mutations in 13 different FALS families.
- Published
- 1993
- Full Text
- View/download PDF
44. Paraoxonase gene mutations in amyotrophic lateral sclerosis
- Author
-
John Landers, Robert H. Brown, Cinzia Gellera, Peter C. Sapp, Diane McKenna-Yasek, Clement E. Furlong, Anne-Marie Wills, Marka van Blitterswijk, Jonathan D. Glass, Ashley Lyn Leclerc, Pamela Keagle, Ildefonso Rodriguez-Leyva, Daryl A. Bosco, Nicola Ticozzi, Antonia Ratti, Franco Taroni, and Vincenzo Silani
- Subjects
DNA Mutational Analysis ,Gene mutation ,medicine.disease_cause ,Article ,Lipid oxidation ,medicine ,Humans ,Family ,Amino Acid Sequence ,Amyotrophic lateral sclerosis ,Gene ,Genetics ,Mutation ,biology ,Sequence Homology, Amino Acid ,Aryldialkylphosphatase ,Amyotrophic Lateral Sclerosis ,Paraoxonase ,Esterases ,medicine.disease ,PON1 ,Neurology ,biology.protein ,Neurology (clinical) - Abstract
Three clustered, homologous paraoxonase genes (PON1, PON2 and PON3) have roles in preventing lipid oxidation and detoxifying organophosphates. Recent reports describe a genetic association between the PON genes and sporadic amyotrophic lateral sclerosis (ALS). We now report that in genomic DNA from individuals with familial and sporadic ALS we have identified at least seven PON gene mutations that are predicted to alter PON function.
- Published
- 2010
45. Analysis of FUS gene mutation in familial amyotrophic lateral sclerosis within an Italian cohort
- Author
-
Robert H. Brown, Nicola Ticozzi, Vincenzo Silani, Peter C. Sapp, Antonia Ratti, Thomas J. Kwiatkowski, Diane McKenna-Yasek, C. Gellera, Franco Taroni, John Landers, Ashley Lyn Leclerc, and Pamela Keagle
- Subjects
Male ,Mutation, Missense ,Biology ,Gene mutation ,medicine.disease_cause ,Cohort Studies ,Exon ,Chromosome 16 ,medicine ,Humans ,Missense mutation ,Genetic Predisposition to Disease ,Genetic Testing ,Amyotrophic lateral sclerosis ,Genetic testing ,Genetics ,Mutation ,Base Sequence ,Models, Genetic ,medicine.diagnostic_test ,Amyotrophic Lateral Sclerosis ,Articles ,Middle Aged ,medicine.disease ,Pedigree ,Italy ,RNA-Binding Protein FUS ,Female ,Neurology (clinical) ,Chromosomes, Human, Pair 16 - Abstract
Objective: Mutations in the FUS gene on chromosome 16 have been recently discovered as a cause of familial amyotrophic lateral sclerosis (FALS). This study determined the frequency and identities of FUS gene mutations in a cohort of Italian patients with FALS. Methods: We screened all 15 coding exons of FUS for mutations in 94 Italian patients with FALS. Results: We identified 4 distinct missense mutations in 5 patients; 2 were novel. The mutations were not present in 376 healthy Italian controls and thus are likely to be pathogenic. Conclusions: Our results demonstrate that FUS mutations cause ∼4% of familial amyotrophic lateral sclerosis cases in the Italian population.
- Published
- 2009
46. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis
- Author
-
Peter C. Sapp, E. J. Kasarskis, Teepu Siddique, A. Davis, Theodore L. Munsat, John Landers, A. L. LeClerc, Betsy A. Hosler, Charles R. Vanderburg, Pietro Cortelli, Robert H. Brown, James M. Gilchrist, Carsten Russ, Guy A. Rouleau, Eric Tamrazian, Diane McKenna-Yasek, Margaret A. Pericak-Vance, H R Horvitz, D. A. Bosco, Jonathan L. Haines, Paul N. Valdmanis, P. J. De Jong, Nicola Ticozzi, Yuko Yoshinaga, Jianhua Yan, Thomas J. Kwiatkowski, Kwiatkowski T.J. Jr., Bosco D.A., Leclerc A.L., Tamrazian E., Vanderburg C.R., Russ C., Davis A., Gilchrist J., Kasarskis E.J., Munsat T., Valdmanis P., Rouleau G.A., Hosler B.A., Cortelli P., de Jong P.J., Yoshinaga Y., Haines J.L., Pericak-Vance M.A., Yan J., Ticozzi N., Siddique T., McKenna-Yasek D., Sapp P.C., Horvitz H.R., Landers J.E., and Brown R.H. Jr.
- Subjects
Male ,Cytoplasm ,Juvenile amyotrophic lateral sclerosis ,Recombinant Fusion Proteins ,Mutation, Missense ,Biology ,TARDBP ,UBQLN2 ,Mice ,C9orf72 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Age of Onset ,Amyotrophic lateral sclerosis ,Cell Nucleus ,Motor Neurons ,Neurons ,Genetics ,Multidisciplinary ,Amyotrophic Lateral Sclerosis ,Brain ,Exons ,Sequence Analysis, DNA ,medicine.disease ,DNA-Binding Proteins ,C9orf72 Protein ,Amino Acid Substitution ,Spinal Cord ,Transportin 1 ,biology.protein ,RNA ,RNA-Binding Protein FUS ,Female ,Mutant Proteins ,Chromosomes, Human, Pair 16 - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal degenerative motor neuron disorder. Ten percent of cases are inherited; most involve unidentified genes. We report here 13 mutations in the fused in sarcoma/translated in liposarcoma ( FUS/TLS ) gene on chromosome 16 that were specific for familial ALS. The FUS/TLS protein binds to RNA, functions in diverse processes, and is normally located predominantly in the nucleus. In contrast, the mutant forms of FUS/TLS accumulated in the cytoplasm of neurons, a pathology that is similar to that of the gene TAR DNA-binding protein 43 ( TDP43 ), whose mutations also cause ALS. Neuronal cytoplasmic protein aggregation and defective RNA metabolism thus appear to be common pathogenic mechanisms involved in ALS and possibly in other neurodegenerative disorders.
- Published
- 2009
47. Linkage of a Gene Causing Familial Amyotrophic Lateral Sclerosis to Chromosome 21 and Evidence of Genetic-Locus Heterogeneity
- Author
-
Teepu Siddique, Denise A. Figlewigz, Margaret A. Pericak-Vance, Jonathan L. Haines, Guy Rouleau, Anita J. Jeffers, Peter Sapp, Wu-Yen Hung, Jacqueline Bebout, Diane McKenna-Yasek, Gang Deng, H. Robert Horvitz, James F. Gusella, Robert H. Brown, and Allen D. Roses
- Subjects
Adult ,Genetic Markers ,Male ,Adolescent ,Chromosomes, Human, Pair 21 ,Genetic Linkage ,Genetic linkage ,Paralysis ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Gene ,Aged ,Lod score ,Linkage (software) ,Genetics ,business.industry ,Amyotrophic Lateral Sclerosis ,Chromosome Mapping ,General Medicine ,Middle Aged ,medicine.disease ,Genetic marker ,Female ,Lod Score ,medicine.symptom ,business ,Chromosome 21 - Abstract
Amyotrophic lateral sclerosis is a progressive neurologic disorder that commonly results in paralysis and death. Despite more than a century of research, no cause of, cure for, or means of preventing this disorder has been found. In a minority of cases, it is familial and inherited as an autosomal dominant trait with age-dependent penetrance. In contrast to the sporadic form of amyotrophic lateral sclerosis, the familial form provides the opportunity to use molecular genetic techniques to localize an inherited defect. Furthermore, such studies have the potential to discover the basic molecular defect causing motor-neuron degeneration.We evaluated 23 families with familial amyotrophic lateral sclerosis for linkage of the gene causing this disease to four DNA markers on the long arm of chromosome 21. Multipoint linkage analyses demonstrated linkage between the gene and these markers. The maximum lod score--5.03--was obtained 10 centimorgans distal (telomeric) to the DNA marker D21S58. There was a significant probability (P less than 0.0001) of genetic-locus heterogeneity in the families.The localization of a gene causing familial amyotrophic lateral sclerosis provides a means of isolating this gene and studying its function. Insight gained from understanding the function of this gene may be applicable to the design of rational therapy for both the familial and sporadic forms of the disease.
- Published
- 1991
- Full Text
- View/download PDF
48. New VAPB deletion variant and exclusion of VAPB mutations in familial ALS
- Author
-
A. F. Henry, T. Cho, Diane McKenna-Yasek, Peter Leigh, Ammar Al-Chalabi, Christopher Shaw, Meraida Polak, A. Virkud, Thomas J. Kwiatkowski, John Landers, Peter C. Sapp, Robert H. Brown, L. Shi, Jonathan D. Glass, M. M. Maxwell, I. Rodriguez-Leyza, and Ashley Lyn Leclerc
- Subjects
Adult ,Central Nervous System ,Genetic Markers ,Male ,Protein Folding ,Genotype ,Recombinant Fusion Proteins ,DNA Mutational Analysis ,Mutation, Missense ,Vesicular Transport Proteins ,Gene mutation ,Biology ,Homology (biology) ,Exon ,Gene Frequency ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Amyotrophic lateral sclerosis ,Gene ,Allele frequency ,Aged ,Genetics ,Amyotrophic Lateral Sclerosis ,VAPB ,Middle Aged ,medicine.disease ,Pedigree ,Amino Acid Substitution ,Female ,Neurology (clinical) ,Gene Deletion ,HeLa Cells - Abstract
Objective: Amyotrophic lateral sclerosis (ALS) is a progressive, neurodegenerative disorder involving upper and lower motor neurons. The vesicle-associated membrane protein B (VAPB) gene has been genetically linked to ALS in several large Brazilian families in which the disorder is caused by a proline to serine mutation at codon 56 (P56S). No additional mutations have been identified. Methods: To establish the prevalence of VAPB mutations, we screened 80 familial ALS samples by DNA sequencing. Results: Our study failed to identify any novel VAPB gene mutations but identified a single Brazilian family harboring the P56S mutation. In a second familial ALS case, we identified a three–base pair deletion within exon 5 of the VAPB gene that deleted the serine residue at position 160 (ΔS160). This variant is detected in a normal population at low frequency (0.45%). Analyses of homology alignment and secondary structure predict that this deletion significantly alters the structure of VAPB, although a GFP-ΔS160 VAPB fusion protein demonstrates a wild-type subcellular localization. This contrasts the aberrant localization observed in a GFP-P56S VAPB fusion protein. The allele frequency of ΔS160 in patients with sporadic ALS does not differ significantly from that in the normal population. Conclusions: Mutations in the VAPB gene are rare and the ΔS160 variant does not contribute to the development of amyotrophic lateral sclerosis.
- Published
- 2008
49. SOD1A4V-mediated ALS: absence of a closely linked modifier gene and origination in Asia
- Author
-
Wendy J. Broom, Peter M. Andersen, Diane McKenna-Yasek, Ammar Al-Chalabi, Carsten Russ, David W. Johnson, Peter C. Sapp, Kristen E. Auwarter, Robert H. Brown, and A J Iafrate
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Asia ,Genotype ,Genetic Linkage ,animal diseases ,SOD1 ,DNA Mutational Analysis ,Single-nucleotide polymorphism ,Locus (genetics) ,Superoxide Dismutase-1 ,Asian People ,medicine ,Coding region ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Amyotrophic lateral sclerosis ,Gene ,Genetics ,Polymorphism, Genetic ,Asian ,Geography ,business.industry ,Superoxide Dismutase ,General Neuroscience ,Haplotype ,Amyotrophic Lateral Sclerosis ,nutritional and metabolic diseases ,Middle Aged ,medicine.disease ,Founder Effect ,nervous system diseases ,Survival Rate ,Phenotype ,nervous system ,Haplotypes ,Mutation ,Female ,business ,Founder effect - Abstract
Familial amyotrophic lateral sclerosis (ALS) accounts for 10% of all ALS. Approximately 20% of cases are due to mutations in the Cu/Zn superoxide dismutase gene (SOD1). In North America, SOD1(A4V) is the most common SOD1 mutation. Carriers of the SOD1(A4V) mutation share a common phenotype with rapid disease progression and death on average occurring at 1.4 years (versus 3-5 years with other dominant SOD1 mutations). Previous studies of SOD1(A4V) carriers identified a common haplotype around the SOD1 locus, suggesting a common founder for most SOD1(A4V) patients. In the current study we sequenced the entire common haplotypic region around SOD1 to test the hypothesis that polymorphisms in either previously undescribed coding regions or non-coding regions around SOD1 are responsible for the more aggressive phenotype in SOD1(A4V)-mediated ALS. We narrowed the conserved region around the SOD1 gene in SOD1(A4V) ALS to 2.8Kb and identified five novel SNPs therein. None of these variants was specifically found in all SOD1(A4V) patients. It therefore appears likely that the aggressive nature of the SOD1(A4V) mutation is not a result of a modifying factor within the region around the SOD1 gene. Founder analysis estimates that the A4V mutation occurred 540 generations (approximately 12,000 years) ago (95% CI 480-700). The conserved minimal haplotype is statistically more similar to Asian than European population DNA sets, suggesting that the A4V mutation arose in native Asian-Americans who reached the Americas through the Bering Strait.
- Published
- 2007
50. Age at onset in sod1-mediated amyotrophic lateral sclerosis shows familiality
- Author
-
Diane McKenna-Yasek, Benjamin M. Neale, Ammar Al-Chalabi, John Powell, Fruhling Rijsdijk, Peter M. Andersen, Robert H. Brown, Pak C. Sham, Joanne Knight, Isabella Fogh, and Vincenzo Silani
- Subjects
medicine.medical_specialty ,Pathology ,Neurology ,SOD1 ,Superoxide dismutase ,Central nervous system disease ,Cellular and Molecular Neuroscience ,Degenerative disease ,Superoxide Dismutase-1 ,Genetics ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Age of Onset ,Motor Neuron Disease ,Genetics (clinical) ,biology ,Models, Genetic ,business.industry ,Superoxide Dismutase ,medicine.disease ,Human genetics ,Founder Effect ,Amino Acid Substitution ,biology.protein ,Age of onset ,business - Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.