1
|
Sabetta E, Ferrari D, Massimo L, Kõks S. Tandem repeat expansions and copy number variations as risk factors and diagnostic tools for amyotrophic lateral sclerosis. Front Neurol 2025; 16:1522445. [PMID: 40012994 PMCID: PMC11860076 DOI: 10.3389/fneur.2025.1522445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disorder leading to upper and lower motoneurons degeneration. Although several mechanisms potentially involved in disease development have been identified, its pathogenesis is not fully understood. From the patient side, ALS diagnosis, still based on clinical criteria, can be difficult and may take up to 1 year. More than 30 genes have been associated to genetically inherited ALS, among which four (C9ORF72, SOD1, TARDBP and FUS) would explain around 60-70% of cases. However, familial ALS represents only 5-10% of ALS cases while the remaining are sporadic, with genetics explaining 6-10% of such cases only. In this context, short tandem repeats (STRs) expansions, have recently been found in clinically diagnosed ALS patients. In this review, we discuss the recent discoveries on ALS associated STRs and their potential as biomarkers as well as prognosis and therapy targets.
Collapse
Affiliation(s)
| | | | | | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Perth, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| |
Collapse
|
2
|
Howard J, Chaouch A, Douglas AGL, MacLeod R, Roggenbuck J, McNeill A. Genetic testing for monogenic forms of motor neuron disease/amyotrophic lateral sclerosis in unaffected family members. Eur J Hum Genet 2025; 33:7-13. [PMID: 39501102 PMCID: PMC11711763 DOI: 10.1038/s41431-024-01718-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 01/11/2025] Open
Abstract
Motor neuron disease (MND), also referred to as amyotrophic lateral sclerosis (ALS), is a monogenic disease in a minority of cases, with autosomal dominant inheritance. Increasing numbers of people with MND are requesting genetic testing, and indeed receiving a genetic diagnosis. Consequently, requests for genetic counselling and predictive testing (i.e. of unaffected family members) are similarly expected to rise, alongside pre-symptomatic clinical trials. Despite this, there is no evidence-based guideline for predictive genetic testing in MND. This paper provides an overview of the genomic basis of MND, focusing specifically on the most common monogenic causes of MND. It then lays out the complexities of MND predictive testing, including the genetic landscape characterised by incomplete penetrance, clinical and genetic heterogeneity, and an oligogenic mechanism of pathogenesis in some cases. Additionally, there is limited research on the psychosocial impact of predictive genetic testing for MND, with studies suggesting potential difficulty in adjusting to the news, in part due to a lack of support and follow-up. This underscores a case for evidence-based, disease-specific guidance for predictive testing in MND.
Collapse
Affiliation(s)
- Jade Howard
- Division of Neuroscience and Neuroscience Institute, The University of Sheffield, Sheffield, UK
| | - Amina Chaouch
- Manchester Centre for Clinical Neuroscience, Salford Royal NHS Foundation Trust, Northern Care Alliance, Stott Lane, M6 8HD, UK
| | - Andrew G L Douglas
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Rhona MacLeod
- Division of Evolution, Infection and Genomics, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jennifer Roggenbuck
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Alisdair McNeill
- Division of Neuroscience and Neuroscience Institute, The University of Sheffield, Sheffield, UK.
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, UK.
| |
Collapse
|
3
|
Grassano M, Canosa A, D’Alfonso S, Corrado L, Brodini G, Koumantakis E, Cugnasco P, Manera U, Vasta R, Palumbo F, Mazzini L, Gallone S, Moglia C, Dewan R, Chia R, Ding J, Dalgard C, Gibbs RJ, Scholz S, Calvo A, Traynor B, Chio A. Intermediate HTT CAG repeats worsen disease severity in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2024; 96:100-102. [PMID: 39242198 PMCID: PMC11672072 DOI: 10.1136/jnnp-2024-333998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/23/2024] [Indexed: 09/09/2024]
Affiliation(s)
- Maurizio Grassano
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
- National Institute on Aging Laboratory of Neurogenetics, Bethesda, Maryland, USA
| | - Antonio Canosa
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| | - Sandra D’Alfonso
- Department of Health Sciences Interdisciplinary Research Center of Autoimmune Diseases, University of Eastern Piedmont Amedeo Avogadro School of Medicine, Novara, Piemonte, Italy
| | - Lucia Corrado
- Department of Health Sciences Interdisciplinary Research Center of Autoimmune Diseases, University of Eastern Piedmont Amedeo Avogadro School of Medicine, Novara, Piemonte, Italy
| | - Giorgia Brodini
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| | | | - Paolo Cugnasco
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| | - Umberto Manera
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
| | - Rosario Vasta
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| | - Francesca Palumbo
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| | - Letizia Mazzini
- University Hospital Maggiore della Carità, Novara, Piemonte, Italy
- University of Eastern Piedmont Amedeo Avogadro School of Medicine, Novara, Piemonte, Italy
| | - Salvatore Gallone
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Piemonte, Italy
| | - Cristina Moglia
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| | - Ramita Dewan
- National Institute on Aging Laboratory of Neurogenetics, Bethesda, Maryland, USA
| | - Ruth Chia
- National Institute on Aging Laboratory of Neurogenetics, Bethesda, Maryland, USA
| | - Jinhui Ding
- National Institute on Aging Laboratory of Neurogenetics, Bethesda, Maryland, USA
| | - Clifton Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Raphael J Gibbs
- National Institute on Aging Laboratory of Neurogenetics, Bethesda, Maryland, USA
| | - Sonja Scholz
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Andrea Calvo
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| | - Bryan Traynor
- National Institute on Aging Laboratory of Neurogenetics, Bethesda, Maryland, USA
- Johns Hopkins University, Baltimore, Maryland, USA
| | - Adriano Chio
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Torino, Piemonte, Italy
| |
Collapse
|
4
|
Bai D, Deng F, Jia Q, Ou K, Wang X, Hou J, Zhu L, Guo M, Yang S, Jiang G, Li S, Li X, Yin P. Pathogenic TDP-43 accelerates the generation of toxic exon1 HTT in Huntington's disease knock-in mice. Aging Cell 2024; 23:e14325. [PMID: 39185703 PMCID: PMC11634733 DOI: 10.1111/acel.14325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
Huntington's disease (HD) is caused by a CAG repeat expansion in exon1 of the HTT gene that encodes a polyglutamine tract in huntingtin protein. The formation of HTT exon1 fragments with an expanded polyglutamine repeat has been implicated as a key step in the pathogenesis of HD. It was reported that the CAG repeat length-dependent aberrant splicing of exon1 HTT results in a short polyadenylated mRNA that is translated into an exon1 HTT protein. Under normal conditions, TDP-43 is predominantly found in the nucleus, where it regulates gene expression. However, in various pathological conditions, TDP-43 is mislocalized in the cytoplasm. By investigating HD knock-in mice, we explore whether the pathogenic TDP-43 in the cytoplasm contributes to HD pathogenesis, through expressing the cytoplasmic TDP-43 without nuclear localization signal. We found that the cytoplasmic TDP-43 is increased in the HD mouse brain and that its mislocalization could deteriorate the motor and gait behavior. Importantly, the cytoplasmic TDP-43, via its binding to the intron1 sequence (GU/UG)n of the mouse Htt pre-mRNA, promotes the transport of exon1-intron1 Htt onto ribosome, resulting in the aberrant generation of exon1 Htt. Our findings suggest that cytoplasmic TDP-43 contributes to HD pathogenesis via its binding to and transport of nuclear un-spliced mRNA to the ribosome for the generation of a toxic protein product.
Collapse
Affiliation(s)
- Dazhang Bai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
- Department of Neurology, Affiliated Hospital of North Sichuan Medical CollegeInstitute of Neurological Diseases, North Sichuan Medical CollegeNanchongSichuanChina
| | - Fuyu Deng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
- Shenzhen Institute for Drug Control, Shenzhen Testing Center of Medical DevicesIn Vitro Diagnostic Reagents Testing DepartmentShenzhenGuangdongChina
| | - Qingqing Jia
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Kaili Ou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Xiang Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Junqi Hou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Longhong Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Mingwei Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Su Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Guohui Jiang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical CollegeInstitute of Neurological Diseases, North Sichuan Medical CollegeNanchongSichuanChina
| | - Shihua Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Xiao‐Jiang Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| | - Peng Yin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Key Laboratory of non‐human Primate Research, Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouGuangdongChina
| |
Collapse
|
5
|
Jiang LL, Zhang XL, Hu HY. Co-Aggregation of TDP-43 with Other Pathogenic Proteins and Their Co-Pathologies in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:12380. [PMID: 39596445 PMCID: PMC11594478 DOI: 10.3390/ijms252212380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Pathological aggregation of a specific protein into insoluble aggregates is a common hallmark of various neurodegenerative diseases (NDDs). In the earlier literature, each NDD is characterized by the aggregation of one or two pathogenic proteins, which can serve as disease-specific biomarkers. The aggregation of these specific proteins is thought to be a major cause of or deleterious result in most NDDs. However, accumulating evidence shows that a pathogenic protein can interact and co-aggregate with other pathogenic proteins in different NDDs, thereby contributing to disease onset and progression synergistically. During the past years, more than one type of NDD has been found to co-exist in some individuals, which may increase the complexity and pathogenicity of these diseases. This article reviews and discusses the biochemical characteristics and molecular mechanisms underlying the co-aggregation and co-pathologies associated with TDP-43 pathology. The TDP-43 aggregates, as a hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), can often be detected in other NDDs, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and spinocerebellar ataxia type 2 (SCA2). In many cases, TDP-43 is shown to interact and co-aggregate with multiple pathogenic proteins in vitro and in vivo. Furthermore, the co-occurrence and co-aggregation of TDP-43 with other pathogenic proteins have important consequences that may aggravate the diseases. Thus, the current viewpoint that the co-aggregation of TDP-43 with other pathogenic proteins in NDDs and their relevance to disease progression may gain insights into the patho-mechanisms and therapeutic potential of various NDDs.
Collapse
Affiliation(s)
- Lei-Lei Jiang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
| | - Xiang-Le Zhang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong-Yu Hu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
| |
Collapse
|
6
|
Saez-Atienzar S, Souza CDS, Chia R, Beal SN, Lorenzini I, Huang R, Levy J, Burciu C, Ding J, Gibbs JR, Jones A, Dewan R, Pensato V, Peverelli S, Corrado L, van Vugt JJFA, van Rheenen W, Tunca C, Bayraktar E, Xia M, Iacoangeli A, Shatunov A, Tiloca C, Ticozzi N, Verde F, Mazzini L, Kenna K, Al Khleifat A, Opie-Martin S, Raggi F, Filosto M, Piccinelli SC, Padovani A, Gagliardi S, Inghilleri M, Ferlini A, Vasta R, Calvo A, Moglia C, Canosa A, Manera U, Grassano M, Mandrioli J, Mora G, Lunetta C, Tanel R, Trojsi F, Cardinali P, Gallone S, Brunetti M, Galimberti D, Serpente M, Fenoglio C, Scarpini E, Comi GP, Corti S, Del Bo R, Ceroni M, Pinter GL, Taroni F, Bella ED, Bersano E, Curtis CJ, Lee SH, Chung R, Patel H, Morrison KE, Cooper-Knock J, Shaw PJ, Breen G, Dobson RJB, Dalgard CL, Scholz SW, Al-Chalabi A, van den Berg LH, McLaughlin R, Hardiman O, Cereda C, Sorarù G, D'Alfonso S, Chandran S, Pal S, Ratti A, Gellera C, Johnson K, Doucet-O'Hare T, Pasternack N, Wang T, Nath A, Siciliano G, Silani V, Başak AN, Veldink JH, Camu W, Glass JD, Landers JE, Chiò A, Sattler R, Shaw CE, Ferraiuolo L, et alSaez-Atienzar S, Souza CDS, Chia R, Beal SN, Lorenzini I, Huang R, Levy J, Burciu C, Ding J, Gibbs JR, Jones A, Dewan R, Pensato V, Peverelli S, Corrado L, van Vugt JJFA, van Rheenen W, Tunca C, Bayraktar E, Xia M, Iacoangeli A, Shatunov A, Tiloca C, Ticozzi N, Verde F, Mazzini L, Kenna K, Al Khleifat A, Opie-Martin S, Raggi F, Filosto M, Piccinelli SC, Padovani A, Gagliardi S, Inghilleri M, Ferlini A, Vasta R, Calvo A, Moglia C, Canosa A, Manera U, Grassano M, Mandrioli J, Mora G, Lunetta C, Tanel R, Trojsi F, Cardinali P, Gallone S, Brunetti M, Galimberti D, Serpente M, Fenoglio C, Scarpini E, Comi GP, Corti S, Del Bo R, Ceroni M, Pinter GL, Taroni F, Bella ED, Bersano E, Curtis CJ, Lee SH, Chung R, Patel H, Morrison KE, Cooper-Knock J, Shaw PJ, Breen G, Dobson RJB, Dalgard CL, Scholz SW, Al-Chalabi A, van den Berg LH, McLaughlin R, Hardiman O, Cereda C, Sorarù G, D'Alfonso S, Chandran S, Pal S, Ratti A, Gellera C, Johnson K, Doucet-O'Hare T, Pasternack N, Wang T, Nath A, Siciliano G, Silani V, Başak AN, Veldink JH, Camu W, Glass JD, Landers JE, Chiò A, Sattler R, Shaw CE, Ferraiuolo L, Fogh I, Traynor BJ. Mechanism-free repurposing of drugs for C9orf72-related ALS/FTD using large-scale genomic data. CELL GENOMICS 2024; 4:100679. [PMID: 39437787 PMCID: PMC11605688 DOI: 10.1016/j.xgen.2024.100679] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 09/22/2024] [Indexed: 10/25/2024]
Abstract
Repeat expansions in the C9orf72 gene are the most common genetic cause of (ALS) and frontotemporal dementia (FTD). Like other genetic forms of neurodegeneration, pinpointing the precise mechanism(s) by which this mutation leads to neuronal death remains elusive, and this lack of knowledge hampers the development of therapy for C9orf72-related disease. We used an agnostic approach based on genomic data (n = 41,273 ALS and healthy samples, and n = 1,516 C9orf72 carriers) to overcome these bottlenecks. Our drug-repurposing screen, based on gene- and expression-pattern matching and information about the genetic variants influencing onset age among C9orf72 carriers, identified acamprosate, a γ-aminobutyric acid analog, as a potentially repurposable treatment for patients carrying C9orf72 repeat expansions. We validated its neuroprotective effect in cell models and showed comparable efficacy to riluzole, the current standard of care. Our work highlights the potential value of genomics in repurposing drugs in situations where the underlying pathomechanisms are inherently complex. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Sara Saez-Atienzar
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Ohio State University, Columbus, OH 43210, USA.
| | - Cleide Dos Santos Souza
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ruth Chia
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Selina N Beal
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ileana Lorenzini
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Jennifer Levy
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Camelia Burciu
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - J Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Ashley Jones
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ramita Dewan
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Viviana Pensato
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Peverelli
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Lucia Corrado
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Joke J F A van Vugt
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ceren Tunca
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Elif Bayraktar
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Cinzia Tiloca
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Letizia Mazzini
- Amyotrophic Lateral Sclerosis Center, Department of Neurology "Maggiore della Carità" University Hospital, Novara, Italy
| | - Kevin Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Sarah Opie-Martin
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Flavia Raggi
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Massimiliano Filosto
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Cotti Piccinelli
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stella Gagliardi
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Maurizio Inghilleri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, 00185 Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Science, University of Ferrara, Ferrara, Italy
| | - Rosario Vasta
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Andrea Calvo
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Cristina Moglia
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Antonio Canosa
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Umberto Manera
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Maurizio Grassano
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Gabriele Mora
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Christian Lunetta
- Department of Neurorehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Institute of Milan, Milan, Italy; NEMO Clinical Center Milano, Fondazione Serena Onlus, Milan, Italy
| | - Raffaella Tanel
- Operative Unit of Neurology, S. Chiara Hospital, Trento, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | | | - Salvatore Gallone
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Maura Brunetti
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Maria Serpente
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Elio Scarpini
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo P Comi
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberto Del Bo
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Ceroni
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Giuseppe Lauria Pinter
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eleonora Dalla Bella
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
| | - Enrica Bersano
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; "L. Sacco" Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Charles J Curtis
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Sang Hyuck Lee
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Raymond Chung
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Hamel Patel
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Karen E Morrison
- School of Medicine, Dentistry, and Biomedical Sciences, Faculty of Medicine Health and Life Sciences, Queen's University, Belfast, UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Gerome Breen
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Richard J B Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK; NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Health Data Research UK London, University College London, London, UK; Institute of Health Informatics, University College London, London, UK; NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, London, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, King's College Hospital, London SE5 9RS, UK
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Russell McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Siddharthan Chandran
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Suvankar Pal
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; Centre for Neuroregeneration and Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Kory Johnson
- Bioinformatics Section, Information Technology Program (ITP), Division of Intramural Research (DIR), National Institute of Neurological Disorders & Stroke, NIH, Bethesda, MD 20892, USA
| | - Tara Doucet-O'Hare
- Neuro-oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Nicholas Pasternack
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Tongguang Wang
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Avindra Nath
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Ayşe Nazlı Başak
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - William Camu
- ALS Center, CHU Gui de Chauliac, University of Montpellier, Montpellier, France
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Rita Sattler
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christopher E Shaw
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Isabella Fogh
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK; National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; RNA Therapeutics Laboratory, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA.
| |
Collapse
|
7
|
Roos AK, Stenvall E, Kockum ES, Grönlund KÅ, Alstermark H, Wuolikainen A, Andersen PM, Nordin A, Forsberg KME. Small striatal huntingtin inclusions in patients with motor neuron disease with reduced penetrance and intermediate HTT gene expansions. Hum Mol Genet 2024; 33:1966-1974. [PMID: 39270726 PMCID: PMC11555821 DOI: 10.1093/hmg/ddae137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Short tandem repeat expansions in the human genome are overrepresented in a variety of neurological disorders. It was recently shown that huntingtin (HTT) repeat expansions with full penetrance, i.e. 40 or more CAG repeats, which normally cause Huntington's disease (HD), are overrepresented in patients with amyotrophic lateral sclerosis (ALS). Whether patients carrying HTT repeat expansions with reduced penetrance, (36-39 CAG repeats), or alleles with intermediate penetrance, (27-35 CAG repeats), have an increased risk of ALS has not yet been investigated. Here, we examined the role of HTT repeat expansions in a motor neuron disease (MND) cohort, searched for expanded HTT alleles, and investigated correlations with phenotype and neuropathology. MND patients harboring C9ORF72 hexanucleotide repeat expansions (HREs) were included, to investigate whether HTT repeat expansions were more common in this group. We found a high prevalence of intermediate (range 5.63%-6.61%) and reduced penetrance (range 0.57%-0.66%) HTT gene expansions in this cohort compared to other populations of European ancestry, but no differences between the MND cohort and the control cohort were observed, regardless of C9ORF72HRE status. Upon autopsy of three patients with intermediate or reduced penetrance HTT alleles, huntingtin inclusions were observed in the caudate nucleus and frontal lobe, but no significant somatic mosaicism was detected in different parts of the nervous system. Thus, we demonstrate, for the first time, huntingtin inclusions in individuals with MND and intermediate and reduced penetrance HTT repeat expansions but more clinicopathological investigations are needed to further understand the impact of HTT gene expansion-related pleiotropy.
Collapse
Affiliation(s)
- Anna-Karin Roos
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Erica Stenvall
- Department of Medical Biosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 2, Umeå SE-90184, Sweden
| | - Emmy Skelton Kockum
- Department of Medical Biosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 2, Umeå SE-90184, Sweden
| | - Kornelia Åman Grönlund
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Helena Alstermark
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Anna Wuolikainen
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala University Hospital, Entrance 85, Floor 2, Uppsala SE-75185, Sweden
| | - Peter M Andersen
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Angelica Nordin
- Department of Medical Biosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 2, Umeå SE-90184, Sweden
| | - Karin M E Forsberg
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| |
Collapse
|
8
|
Luan T, Li Q, Huang Z, Feng Y, Xu D, Zhou Y, Hu Y, Wang T. Axonopathy Underlying Amyotrophic Lateral Sclerosis: Unraveling Complex Pathways and Therapeutic Insights. Neurosci Bull 2024; 40:1789-1810. [PMID: 39097850 PMCID: PMC11607281 DOI: 10.1007/s12264-024-01267-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/08/2024] [Indexed: 08/05/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disorder characterized by progressive axonopathy, jointly leading to the dying back of the motor neuron, disrupting both nerve signaling and motor control. In this review, we highlight the roles of axonopathy in ALS progression, driven by the interplay of multiple factors including defective trafficking machinery, protein aggregation, and mitochondrial dysfunction. Dysfunctional intracellular transport, caused by disruptions in microtubules, molecular motors, and adaptors, has been identified as a key contributor to disease progression. Aberrant protein aggregation involving TDP-43, FUS, SOD1, and dipeptide repeat proteins further amplifies neuronal toxicity. Mitochondrial defects lead to ATP depletion, oxidative stress, and Ca2+ imbalance, which are regarded as key factors underlying the loss of neuromuscular junctions and axonopathy. Mitigating these defects through interventions including neurotrophic treatments offers therapeutic potential. Collaborative research efforts aim to unravel ALS complexities, opening avenues for holistic interventions that target diverse pathological mechanisms.
Collapse
Affiliation(s)
- Tongshu Luan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Qing Li
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Zhi Huang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yu Feng
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Duo Xu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yujie Zhou
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yiqing Hu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Tong Wang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
9
|
George G, Ajayan A, Varkey J, Pandey NK, Chen J, Langen R. TDP43 and huntingtin Exon-1 undergo a conformationally specific interaction that strongly alters the fibril formation of both proteins. J Biol Chem 2024; 300:107660. [PMID: 39128727 PMCID: PMC11408864 DOI: 10.1016/j.jbc.2024.107660] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024] Open
Abstract
Protein aggregation is a common feature of many neurodegenerative diseases. In Huntington's disease, mutant huntingtin is the primary aggregating protein, but the aggregation of other proteins, such as TDP43, is likely to further contribute to toxicity. Moreover, mutant huntingtin is also a risk factor for TDP pathology in ALS. Despite this co-pathology of huntingtin and TDP43, it remains unknown whether these amyloidogenic proteins directly interact with each other. Using a combination of biophysical methods, we show that the aggregation-prone regions of both proteins, huntingtin exon-1 (Httex1) and the TDP43 low complexity domain (TDP43-LCD), interact in a conformationally specific manner. This interaction significantly slows Httex1 aggregation, while it accelerates TDP43-LCD aggregation. A key intermediate responsible for both effects is a complex formed by liquid TDP43-LCD condensates and Httex1 fibrils. This complex shields seeding competent surfaces of Httex1 fibrils from Httex1 monomers, which are excluded from the condensates. In contrast, TDP43-LCD condensates undergo an accelerated liquid-to-solid transition upon exposure to Httex1 fibrils. Cellular studies show co-aggregation of untagged Httex1 with TDP43. This interaction causes mislocalization of TDP43, which has been linked to TDP43 toxicity. The protection from Httex1 aggregation in lieu of TDP43-LCD aggregation is interesting, as it mirrors what has been found in disease models, namely that TDP43 can protect from huntingtin toxicity, while mutant huntingtin can promote TDP43 pathology. These results suggest that direct protein interaction could, at least in part, be responsible for the linked pathologies of both proteins.
Collapse
Affiliation(s)
- Gincy George
- Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Anakha Ajayan
- Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jobin Varkey
- Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nitin K Pandey
- Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jeannie Chen
- Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ralf Langen
- Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
10
|
Chia R, Ray A, Shah Z, Ding J, Ruffo P, Fujita M, Menon V, Saez-Atienzar S, Reho P, Kaivola K, Walton RL, Reynolds RH, Karra R, Sait S, Akcimen F, Diez-Fairen M, Alvarez I, Fanciulli A, Stefanova N, Seppi K, Duerr S, Leys F, Krismer F, Sidoroff V, Zimprich A, Pirker W, Rascol O, Foubert-Samier A, Meissner WG, Tison F, Pavy-Le Traon A, Pellecchia MT, Barone P, Russillo MC, Marín-Lahoz J, Kulisevsky J, Torres S, Mir P, Periñán MT, Proukakis C, Chelban V, Wu L, Goh YY, Parkkinen L, Hu MT, Kobylecki C, Saxon JA, Rollinson S, Garland E, Biaggioni I, Litvan I, Rubio I, Alcalay RN, Kwei KT, Lubbe SJ, Mao Q, Flanagan ME, Castellani RJ, Khurana V, Ndayisaba A, Calvo A, Mora G, Canosa A, Floris G, Bohannan RC, Moore A, Norcliffe-Kaufmann L, Palma JA, Kaufmann H, Kim C, Iba M, Masliah E, Dawson TM, Rosenthal LS, Pantelyat A, Albert MS, Pletnikova O, Troncoso JC, Infante J, Lage C, Sánchez-Juan P, Serrano GE, Beach TG, Pastor P, Morris HR, Albani D, Clarimon J, Wenning GK, Hardy JA, Ryten M, Topol E, Torkamani A, Chiò A, Bennett DA, De Jager PL, Low PA, Singer W, Cheshire WP, Wszolek ZK, Dickson DW, et alChia R, Ray A, Shah Z, Ding J, Ruffo P, Fujita M, Menon V, Saez-Atienzar S, Reho P, Kaivola K, Walton RL, Reynolds RH, Karra R, Sait S, Akcimen F, Diez-Fairen M, Alvarez I, Fanciulli A, Stefanova N, Seppi K, Duerr S, Leys F, Krismer F, Sidoroff V, Zimprich A, Pirker W, Rascol O, Foubert-Samier A, Meissner WG, Tison F, Pavy-Le Traon A, Pellecchia MT, Barone P, Russillo MC, Marín-Lahoz J, Kulisevsky J, Torres S, Mir P, Periñán MT, Proukakis C, Chelban V, Wu L, Goh YY, Parkkinen L, Hu MT, Kobylecki C, Saxon JA, Rollinson S, Garland E, Biaggioni I, Litvan I, Rubio I, Alcalay RN, Kwei KT, Lubbe SJ, Mao Q, Flanagan ME, Castellani RJ, Khurana V, Ndayisaba A, Calvo A, Mora G, Canosa A, Floris G, Bohannan RC, Moore A, Norcliffe-Kaufmann L, Palma JA, Kaufmann H, Kim C, Iba M, Masliah E, Dawson TM, Rosenthal LS, Pantelyat A, Albert MS, Pletnikova O, Troncoso JC, Infante J, Lage C, Sánchez-Juan P, Serrano GE, Beach TG, Pastor P, Morris HR, Albani D, Clarimon J, Wenning GK, Hardy JA, Ryten M, Topol E, Torkamani A, Chiò A, Bennett DA, De Jager PL, Low PA, Singer W, Cheshire WP, Wszolek ZK, Dickson DW, Traynor BJ, Gibbs JR, Dalgard CL, Ross OA, Houlden H, Scholz SW. Genome sequence analyses identify novel risk loci for multiple system atrophy. Neuron 2024; 112:2142-2156.e5. [PMID: 38701790 PMCID: PMC11223971 DOI: 10.1016/j.neuron.2024.04.002] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/28/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024]
Abstract
Multiple system atrophy (MSA) is an adult-onset, sporadic synucleinopathy characterized by parkinsonism, cerebellar ataxia, and dysautonomia. The genetic architecture of MSA is poorly understood, and treatments are limited to supportive measures. Here, we performed a comprehensive analysis of whole genome sequence data from 888 European-ancestry MSA cases and 7,128 controls to systematically investigate the genetic underpinnings of this understudied neurodegenerative disease. We identified four significantly associated risk loci using a genome-wide association study approach. Transcriptome-wide association analyses prioritized USP38-DT, KCTD7, and lnc-KCTD7-2 as novel susceptibility genes for MSA within these loci, and single-nucleus RNA sequence analysis found that the associated variants acted as cis-expression quantitative trait loci for multiple genes across neuronal and glial cell types. In conclusion, this study highlights the role of genetic determinants in the pathogenesis of MSA, and the publicly available data from this study represent a valuable resource for investigating synucleinopathies.
Collapse
Affiliation(s)
- Ruth Chia
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Anindita Ray
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Zalak Shah
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Paola Ruffo
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA; Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Masashi Fujita
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Sara Saez-Atienzar
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Paolo Reho
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Karri Kaivola
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ronald L Walton
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Regina H Reynolds
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK; Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Ramita Karra
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Shaimaa Sait
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Fulya Akcimen
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Monica Diez-Fairen
- Memory and Movement Disorders Units, Department of Neurology, University Hospital Mutua de Terrassa, Barcelona, Spain
| | - Ignacio Alvarez
- Memory and Movement Disorders Units, Department of Neurology, University Hospital Mutua de Terrassa, Barcelona, Spain
| | | | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Susanne Duerr
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Fabian Leys
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krismer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Victoria Sidoroff
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Walter Pirker
- Department of Neurology, Klinik Ottakring - Wilhelminenspital, Vienna, Austria
| | - Olivier Rascol
- MSA French Reference Center and CIC-1436, Department of Clinical Pharmacology and Neurosciences, University of Toulouse, Toulouse, France
| | - Alexandra Foubert-Samier
- Service de Neurologie des Maladies Neurodégénératives, French Reference Center for MSA, NS-Park/FCRIN Network, CHU Bordeaux, Bordeaux, France
| | - Wassilios G Meissner
- Service de Neurologie des Maladies Neurodégénératives, French Reference Center for MSA, NS-Park/FCRIN Network, CHU Bordeaux, Bordeaux, France; University of Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, France; Department of Medicine, University of Otago, and the New Zealand Brain Research Institute, Christchurch, New Zealand
| | - François Tison
- Service de Neurologie des Maladies Neurodégénératives, French Reference Center for MSA, NS-Park/FCRIN Network, CHU Bordeaux, Bordeaux, France; University of Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, France
| | - Anne Pavy-Le Traon
- French Reference Center for MSA, Department of Neurosciences, Centre d'Investigation Clinique de Toulouse CIC1436, UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University Hospital of Toulouse, INSERM, Toulouse, France
| | - Maria Teresa Pellecchia
- Neuroscience Section, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Paolo Barone
- Neuroscience Section, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Maria Claudia Russillo
- Neuroscience Section, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Juan Marín-Lahoz
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Institut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau), Centro de Investigación en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain; Servicio de Neurología, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Institut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau), Centro de Investigación en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Soraya Torres
- Institut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau), Centro de Investigación en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Departamento de Medicina Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Maria Teresa Periñán
- Unidad de Trastornos del Movimiento Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain; Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University, London, UK
| | - Christos Proukakis
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK
| | - Viorica Chelban
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK; The National Hospital for Neurology and Neurosurgery, London, UK
| | - Lesley Wu
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK
| | - Yee Y Goh
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK
| | - Laura Parkkinen
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Michele T Hu
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Christopher Kobylecki
- Department of Neurology, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, UK
| | - Jennifer A Saxon
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Salfort, UK; Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Sara Rollinson
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Emily Garland
- Autonomic Dysfunction Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Italo Biaggioni
- Autonomic Dysfunction Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Irene Litvan
- Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Ileana Rubio
- Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Kimberly T Kwei
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Steven J Lubbe
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qinwen Mao
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA; Department of Pathology, UT Health San Antonio, San Antonio, TX, USA
| | - Rudolph J Castellani
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Vikram Khurana
- Ann Romney Center for Neurologic Disease, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Alain Ndayisaba
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria; Ann Romney Center for Neurologic Disease, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrea Calvo
- "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Gabriele Mora
- Istituti Clinici Scientifici Maugeri, IRCCS, Milan, Italy
| | - Antonio Canosa
- "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Gianluca Floris
- Department of Neurology, University Hospital of Cagliari, Cagliari, Italy
| | - Ryan C Bohannan
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Anni Moore
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | | | - Jose-Alberto Palma
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | - Horacio Kaufmann
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | - Changyoun Kim
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Michiyo Iba
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Eliezer Masliah
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Ted M Dawson
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA; Neuroregeneration and Stem Cell Programs, Institute of Cell Engineering, Johns Hopkins University Medical Center, Baltimore, MD, USA; Department of Pharmacology and Molecular Science, Johns Hopkins University Medical Center, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Liana S Rosenthal
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Alexander Pantelyat
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Olga Pletnikova
- Department of Pathology (Neuropathology), Johns Hopkins University Medical Center, Baltimore, MD, USA; Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Juan C Troncoso
- Department of Pathology (Neuropathology), Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Jon Infante
- Neurology Service, University Hospital Marqués de Valdecilla-IDIVAL-UC-CIBERNED, Santander, Spain
| | - Carmen Lage
- Neurology Service, University Hospital Marqués de Valdecilla-IDIVAL-UC-CIBERNED, Santander, Spain
| | - Pascual Sánchez-Juan
- Neurology Service, University Hospital Marqués de Valdecilla-IDIVAL-UC-CIBERNED, Santander, Spain; Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, Madrid, Spain
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Pau Pastor
- Genomics and Transcriptomics of Synucleinopathies, Neurosciences, The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain; Unit of Neurodegenerative Diseases, Department of Neurology, University Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Huw R Morris
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jordi Clarimon
- Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; The Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Gregor K Wenning
- Autonomic Unit - Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - John A Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute of UCL, UCL Institute of Neurology, University College London, London, UK; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London, UK; UCL Movement Disorders Centre, University College London, London, UK; Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Mina Ryten
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK; Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
| | - Eric Topol
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA, USA
| | - Ali Torkamani
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA, USA
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Philip A Low
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA; RNA Therapeutics Laboratory, Therapeutics Development Branch, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - J Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, USA
| | - Henry Houlden
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK; The National Hospital for Neurology and Neurosurgery, London, UK
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA.
| |
Collapse
|
11
|
Pérez‐Oliveira S, Castilla‐Silgado J, Painous C, Aldecoa I, Menéndez‐González M, Blázquez‐Estrada M, Corte D, Tomás‐Zapico C, Compta Y, Muñoz E, Lladó A, Balasa M, Aragonès G, García‐González P, Rosende‐Roca M, Boada M, Ruíz A, Pastor P, De la Casa‐Fages B, Rabano A, Sánchez‐Valle R, Molina‐Porcel L, Álvarez V. Huntingtin CAG repeats in neuropathologically confirmed tauopathies: Novel insights. Brain Pathol 2024; 34:e13250. [PMID: 38418081 PMCID: PMC11189778 DOI: 10.1111/bpa.13250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/09/2024] [Indexed: 03/01/2024] Open
Abstract
Previous studies have suggested a relationship between the number of CAG triplet repeats in the HTT gene and neurodegenerative diseases not related to Huntington's disease (HD). This study seeks to investigate whether the number of CAG repeats of HTT is associated with the risk of developing certain tauopathies and its influence as a modulator of the clinical and neuropathological phenotype. Additionally, it aims to evaluate the potential of polyglutamine staining as a neuropathological screening. We genotyped the HTT gene CAG repeat number and APOE-ℰ isoforms in a cohort of patients with neuropathological diagnoses of tauopathies (n=588), including 34 corticobasal degeneration (CBD), 98 progressive supranuclear palsy (PSP) and 456 Alzheimer's disease (AD). Furthermore, we genotyped a control group of 1070 patients, of whom 44 were neuropathologic controls. We identified significant differences in the number of patients with pathological HTT expansions in the CBD group (2.7%) and PSP group (3.2%) compared to control subjects (0.2%). A significant increase in the size of the HTT CAG repeats was found in the AD compared to the control group, influenced by the presence of the Apoliprotein E (APOE)-ℰ4 isoform. Post-mortem assessments uncovered tauopathy pathology with positive polyglutamine aggregates, with a slight predominance in the neostriatum for PSP and CBD cases and somewhat greater limbic involvement in the AD case. Our results indicated a link between HTT CAG repeat expansion with other non-HD pathology, suggesting they could share common neurodegenerative pathways. These findings support that genetic or histological screening for HTT repeat expansions should be considered in tauopathies.
Collapse
Affiliation(s)
- Sergio Pérez‐Oliveira
- Laboratory of GeneticsHospital Universitario Central de AsturiasOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Juan Castilla‐Silgado
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
- Department of Functional Biology (Physiology)University of OviedoOviedoSpain
| | - Cèlia Painous
- Parkinson's Disease and Movement Disorders Unit, Department of NeurologyHospital Clinic of BarcelonaBarcelonaSpain
- UB Neuro Institut de Neurociències, Maeztu CenterUniversity of BarcelonaBarcelonaSpain
- Fundació de Recerca Clínic Barcelona‐Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB‐IDIBAPS)BarcelonaSpain
| | - Iban Aldecoa
- Neurological Tissue Bank of the Biobank‐Hospital Clinic‐FRCB‐IDIBAPSBarcelonaSpain
- Pathology Department, Biomedical Diagnostic CenterHospital Clínic de Barcelona, University of BarcelonaBarcelonaSpain
| | - Manuel Menéndez‐González
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
- Department of NeurologyHospital Universitario Central de AsturiasOviedoSpain
- Department of MedicineUniversity of OviedoOviedoSpain
| | - Marta Blázquez‐Estrada
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
- Department of NeurologyHospital Universitario Central de AsturiasOviedoSpain
- Department of MedicineUniversity of OviedoOviedoSpain
| | - Daniela Corte
- Biobank of Principado de Asturias, Hospital Universitario Central de Asturias (HUCA)OviedoSpain
| | - Cristina Tomás‐Zapico
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
- Department of Functional Biology (Physiology)University of OviedoOviedoSpain
| | - Yaroslau Compta
- Parkinson's Disease and Movement Disorders Unit, Department of NeurologyHospital Clinic of BarcelonaBarcelonaSpain
- UB Neuro Institut de Neurociències, Maeztu CenterUniversity of BarcelonaBarcelonaSpain
- Fundació de Recerca Clínic Barcelona‐Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB‐IDIBAPS)BarcelonaSpain
| | - Esteban Muñoz
- Parkinson's Disease and Movement Disorders Unit, Department of NeurologyHospital Clinic of BarcelonaBarcelonaSpain
- UB Neuro Institut de Neurociències, Maeztu CenterUniversity of BarcelonaBarcelonaSpain
- Fundació de Recerca Clínic Barcelona‐Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB‐IDIBAPS)BarcelonaSpain
| | - Albert Lladó
- Alzheimer's Disease and other Cognitive Disorders UnitNeurology Service, Hospital Clínic, FRCB‐IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Mircea Balasa
- Alzheimer's Disease and other Cognitive Disorders UnitNeurology Service, Hospital Clínic, FRCB‐IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Gemma Aragonès
- Neurological Tissue Bank of the Biobank‐Hospital Clinic‐FRCB‐IDIBAPSBarcelonaSpain
| | - Pablo García‐González
- Ace Alzheimer Center Barcelona – Universitat Internacional de CatalunyaBarcelonaSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Maitée Rosende‐Roca
- Ace Alzheimer Center Barcelona – Universitat Internacional de CatalunyaBarcelonaSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de CatalunyaBarcelonaSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Agustín Ruíz
- Ace Alzheimer Center Barcelona – Universitat Internacional de CatalunyaBarcelonaSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Pau Pastor
- Unit of Neurodegenerative Diseases, Department of NeurologyUniversity Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) BadalonaBarcelonaSpain
| | - Beatriz De la Casa‐Fages
- Movement Disorders Unit, Department of NeurologyHospital General Universitario Gregorio MarañónMadridSpain
- Instituto Investigación Sanitaria Gregorio MarañónMadridSpain
| | - Alberto Rabano
- Neuropathology Department and Brain Tissue BankCIEN Foundation, Queen Sofia Foundation Alzheimer CenterMadridSpain
| | - Raquel Sánchez‐Valle
- Alzheimer's Disease and other Cognitive Disorders UnitNeurology Service, Hospital Clínic, FRCB‐IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Laura Molina‐Porcel
- UB Neuro Institut de Neurociències, Maeztu CenterUniversity of BarcelonaBarcelonaSpain
- Alzheimer's Disease and other Cognitive Disorders UnitNeurology Service, Hospital Clínic, FRCB‐IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Victoria Álvarez
- Laboratory of GeneticsHospital Universitario Central de AsturiasOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| |
Collapse
|
12
|
Ruiz de Sabando A, Ciosi M, Galbete A, Cumming SA, Monckton DG, Ramos-Arroyo MA. Somatic CAG repeat instability in intermediate alleles of the HTT gene and its potential association with a clinical phenotype. Eur J Hum Genet 2024; 32:770-778. [PMID: 38433266 PMCID: PMC11220145 DOI: 10.1038/s41431-024-01546-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 03/05/2024] Open
Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by ≥36 CAGs in the HTT gene. Intermediate alleles (IAs) (27-35 CAGs) are not considered HD-causing, but their potential association with neurocognitive symptoms remains controversial. As HTT somatic CAG expansion influences HD onset, we hypothesised that IAs are somatically unstable, and that somatic CAG expansion may drive phenotypic presentation in some IA carriers. We quantified HTT somatic CAG expansions by MiSeq sequencing in the blood DNA of 164 HD subjects and 191 IA (symptomatic and control) carriers, and in the brain DNA of a symptomatic 33 CAG carrier. We also performed genotype-phenotype analysis. The phenotype of symptomatic IA carriers was characterised by motor (85%), cognitive (27%) and/or behavioural (29%) signs, with a late (58.7 ± 18.6 years), but not CAG-dependent, age at onset. IAs displayed somatic expansion that were CAG and age-dependent in blood DNA, with 0.4% and 0.01% of DNA molecules expanding by CAG and year, respectively. Somatic expansions of +1 and +2 CAGs were detected in the brain of the individual with 33 CAGs, with the highest expansion frequency in the putamen (10.3%) and the lowest in the cerebellum (4.8%). Somatic expansion in blood DNA was not different in symptomatic vs. control IA carriers. In conclusion, we show that HTT IAs are somatically unstable, but we found no association with HD-like phenotypes. It is plausible, however, that some IAs, close to the HD pathological threshold and with a predisposing genetic background, could manifest with neurocognitive symptoms.
Collapse
Affiliation(s)
- Ainara Ruiz de Sabando
- Department of Medical Genetics, Hospital Universitario de Navarra, IdiSNA, 31008, Pamplona, Spain
- Department of Health Sciences, Universidad Pública de Navarra, IdiSNA, 31008, Pamplona, Spain
- Fundación Miguel Servet-Navarrabiomed, IdiSNA, 31008, Pamplona, Spain
| | - Marc Ciosi
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Arkaitz Galbete
- Department of Statistics, Informatics and Mathematics, Universidad Pública de Navarra, IdiSNA, 31006, Pamplona, Spain
| | - Sarah A Cumming
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Darren G Monckton
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Maria A Ramos-Arroyo
- Department of Medical Genetics, Hospital Universitario de Navarra, IdiSNA, 31008, Pamplona, Spain.
- Fundación Miguel Servet-Navarrabiomed, IdiSNA, 31008, Pamplona, Spain.
| |
Collapse
|
13
|
Sellier C, Corcia P, Vourc'h P, Dupuis L. C9ORF72 hexanucleotide repeat expansion: From ALS and FTD to a broader pathogenic role? Rev Neurol (Paris) 2024; 180:417-428. [PMID: 38609750 DOI: 10.1016/j.neurol.2024.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024]
Abstract
The major gene underlying monogenic forms of amyotrophic lateral sclerosis (ALS) and fronto-temporal dementia (FTD) is C9ORF72. The causative mutation in C9ORF72 is an abnormal hexanucleotide (G4C2) repeat expansion (HRE) located in the first intron of the gene. The aim of this review is to propose a comprehensive update on recent developments on clinical, biological and therapeutics aspects related to C9ORF72 in order to highlight the current understanding of genotype-phenotype correlations, and also on biological machinery leading to neuronal death. We will particularly focus on the broad phenotypic presentation of C9ORF72-related diseases, that goes well beyond the classical phenotypes observed in ALS and FTD patients. Last, we will comment the possible therapeutical hopes for patients carrying a C9ORF72 HRE.
Collapse
Affiliation(s)
- C Sellier
- Centre de recherches en biomédecine de Strasbourg, UMR-S1329, Inserm, université de Strasbourg, Strasbourg, France
| | - P Corcia
- UMR 1253 iBrain, Inserm, université de Tours, Tours, France; Centre constitutif de coordination SLA, CHU de Bretonneau, 2, boulevard Tonnelle, 37044 Tours cedex 1, France
| | - P Vourc'h
- UMR 1253 iBrain, Inserm, université de Tours, Tours, France; Service de biochimie et biologie moléculaire, CHU de Tours, Tours, France
| | - L Dupuis
- Centre de recherches en biomédecine de Strasbourg, UMR-S1329, Inserm, université de Strasbourg, Strasbourg, France.
| |
Collapse
|
14
|
Canosa A, Cabras S, Di Pede F, Manera U, Vasta R, Moglia C, Calvo A, Gallone S, Chiò A. A mother and her daughter carrying a pathogenic expansion of the HTT gene with a phenotype encompassing motor neuron disease and Huntington's disease. Clin Genet 2024; 105:430-433. [PMID: 38092667 DOI: 10.1111/cge.14472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 03/07/2024]
Abstract
Recently, pathogenic expansions (range 40-64 CAG repeats) in the HTT gene have been found in patients diagnosed with pure frontotemporal dementia/amyotrophic lateral sclerosis (FTD/ALS). We report a mother with Huntington's disease (HD) associated with motor neuron disease (MND) signs and her daughter suffering from ALS with subtle signs of HD, both carrying a pathogenic allele of the HTT gene (i.e., >39 repeats). The co-occurrence of MND and chorea has been reported in previous cases. Subjects showing both ALS and HD signs and carrying HTT pathogenic expansions in two generations of the same kindred have never been reported so far. The study of the overlap of disease mechanisms at the cellular level between TDP-43 and Huntingtin is relevant in an era offering promising strategies of targeted treatments in neurodegenerative disorders.
Collapse
Affiliation(s)
- Antonio Canosa
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
- Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy
| | - Sara Cabras
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- University of Camerino, Centre for Neuroscience, Camerino, Italy
| | - Francesca Di Pede
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Umberto Manera
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Rosario Vasta
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Cristina Moglia
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Andrea Calvo
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
- Neuroscience Institute of Turin (NIT), Turin, Italy
| | - Salvatore Gallone
- SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Adriano Chiò
- ALS Centre, 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
- SC Neurologia 1U, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
- Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy
- Neuroscience Institute of Turin (NIT), Turin, Italy
| |
Collapse
|
15
|
Novy C, Busk ØL, Tysnes OB, Landa SS, Aanjesen TN, Alstadhaug KB, Bjerknes TL, Bjørnå IK, Bråthen G, Dahl E, Demic N, Fahlström M, Flemmen HØ, Hallerstig E, HogenEsch I, Kampman MT, Kleveland G, Kvernmo HB, Ljøstad U, Maniaol A, Morsund AH, Nakken O, Olsen CG, Schlüter K, Utvik MS, Yaseen R, Holla ØL, Holmøy T, Høyer H. Repeat expansions in AR, ATXN1, ATXN2 and HTT in Norwegian patients diagnosed with amyotrophic lateral sclerosis. Brain Commun 2024; 6:fcae087. [PMID: 38585669 PMCID: PMC10998343 DOI: 10.1093/braincomms/fcae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/23/2024] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
Genetic repeat expansions cause neuronal degeneration in amyotrophic lateral sclerosis as well as other neurodegenerative disorders such as spinocerebellar ataxia, Huntington's disease and Kennedy's disease. Repeat expansions in the same gene can cause multiple clinical phenotypes. We aimed to characterize repeat expansions in a Norwegian amyotrophic lateral sclerosis cohort. Norwegian amyotrophic lateral sclerosis patients (n = 414) and neurologically healthy controls adjusted for age and gender (n = 713) were investigated for repeat expansions in AR, ATXN1, ATXN2 and HTT using short read exome sequencing and the ExpansionHunter software. Five amyotrophic lateral sclerosis patients (1.2%) and two controls (0.3%) carried ≥36 repeats in HTT (P = 0.032), and seven amyotrophic lateral sclerosis patients (1.7%) and three controls (0.4%) carried ≥29 repeats in ATXN2 (P = 0.038). One male diagnosed with amyotrophic lateral sclerosis carried a pathogenic repeat expansion in AR, and his diagnosis was revised to Kennedy's disease. In ATXN1, 50 amyotrophic lateral sclerosis patients (12.1%) and 96 controls (13.5%) carried ≥33 repeats (P = 0.753). None of the patients with repeat expansions in ATXN2 or HTT had signs of Huntington's disease or spinocerebellar ataxia type 2, based on a re-evaluation of medical records. The diagnosis of amyotrophic lateral sclerosis was confirmed in all patients, with the exception of one patient who had primary lateral sclerosis. Our findings indicate that repeat expansions in HTT and ATXN2 are associated with increased likelihood of developing amyotrophic lateral sclerosis. Further studies are required to investigate the potential relationship between HTT repeat expansions and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Camilla Novy
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Øyvind L Busk
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Ole-Bjørn Tysnes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5009 Bergen, Norway
| | - Sigve S Landa
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Tori N Aanjesen
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
| | | | - Tale L Bjerknes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5009 Bergen, Norway
- Institute of Clinical Medicine, University of Bergen, 5007 Bergen, Norway
| | - Ingrid K Bjørnå
- Department of Neurology, Vestre Viken Hospital Trust, 3004 Drammen, Norway
| | - Geir Bråthen
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Elin Dahl
- Department of Neurology, Telemark Hospital Trust, 3710 Skien, Norway
| | - Natasha Demic
- Department of Neurology, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| | - Maria Fahlström
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Heidi Ø Flemmen
- Department of Neurology, Telemark Hospital Trust, 3710 Skien, Norway
| | - Erika Hallerstig
- Department of Neurology, Østfold Hospital Trust, 1714 Grålum, Norway
| | - Ineke HogenEsch
- Department of Neurology, Fonna Hospital Trust, 5528 Haugesund, Norway
| | - Margitta T Kampman
- Department of Neurology, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Grethe Kleveland
- Department of Neurology, Innlandet Hospital Trust, 2609 Lillehammer, Norway
| | - Helene B Kvernmo
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Unn Ljøstad
- Institute of Clinical Medicine, University of Bergen, 5007 Bergen, Norway
- Department of Neurology, Sørlandet Hospital Trust, 4615 Kristiansand, Norway
| | - Angelina Maniaol
- Department of Neurology, Oslo University Hospital, 0450 Oslo, Norway
| | | | - Ola Nakken
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Cathrine G Olsen
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Katrin Schlüter
- Department of Neurology, Stavanger University Hospital, 4019 Stavanger, Norway
| | - May-Sissel Utvik
- Department of Neurology, Namsos Hospital Trust, 7803 Namsos, Norway
| | - Ryaz Yaseen
- Department of Neurology, Oslo University Hospital, 0450 Oslo, Norway
| | - Øystein L Holla
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Trygve Holmøy
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Helle Høyer
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| |
Collapse
|
16
|
Prange S, Laurencin C, Roche P, Quadrio I, Thobois S. PSP-Richardson's Syndrome as a Rare Phenotypic Expression of Very Late-Onset Huntington's Disease: A Case Report. Mov Disord Clin Pract 2024; 11:303-305. [PMID: 38173343 PMCID: PMC10928328 DOI: 10.1002/mdc3.13943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/09/2023] [Accepted: 11/10/2023] [Indexed: 01/05/2024] Open
Affiliation(s)
- Stephane Prange
- Hospices Civils de Lyon, Department of Neurology C, Expert Parkinson Center NS‐PARK/FCRINPierre Wertheimer Neurological HospitalBronFrance
- Univ Lyon, Marc Jeannerod Cognitive Neuroscience Institute, CNRS, UMR 5229BronFrance
- Univ Lyon, Faculté de Médecine et de Maïeutique Lyon Sud Charles MérieuxUniversité Claude Bernard Lyon 1OullinsFrance
| | - Chloé Laurencin
- Hospices Civils de Lyon, Department of Neurology C, Expert Parkinson Center NS‐PARK/FCRINPierre Wertheimer Neurological HospitalBronFrance
| | - Pauline Roche
- Hospices Civils de Lyon, Department of Neurology C, Expert Parkinson Center NS‐PARK/FCRINPierre Wertheimer Neurological HospitalBronFrance
| | - Isabelle Quadrio
- Hospices Civils de Lyon, Neurobiology and NeurogeneticsDepartment of Biochemistry and Molecular BiologyLyonFrance
| | - Stéphane Thobois
- Hospices Civils de Lyon, Department of Neurology C, Expert Parkinson Center NS‐PARK/FCRINPierre Wertheimer Neurological HospitalBronFrance
- Univ Lyon, Marc Jeannerod Cognitive Neuroscience Institute, CNRS, UMR 5229BronFrance
- Univ Lyon, Faculté de Médecine et de Maïeutique Lyon Sud Charles MérieuxUniversité Claude Bernard Lyon 1OullinsFrance
| |
Collapse
|
17
|
Rizzuti M, Sali L, Melzi V, Scarcella S, Costamagna G, Ottoboni L, Quetti L, Brambilla L, Papadimitriou D, Verde F, Ratti A, Ticozzi N, Comi GP, Corti S, Gagliardi D. Genomic and transcriptomic advances in amyotrophic lateral sclerosis. Ageing Res Rev 2023; 92:102126. [PMID: 37972860 DOI: 10.1016/j.arr.2023.102126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder and the most common motor neuron disease. ALS shows substantial clinical and molecular heterogeneity. In vitro and in vivo models coupled with multiomic techniques have provided important contributions to unraveling the pathomechanisms underlying ALS. To date, despite promising results and accumulating knowledge, an effective treatment is still lacking. Here, we provide an overview of the literature on the use of genomics, epigenomics, transcriptomics and microRNAs to deeply investigate the molecular mechanisms developing and sustaining ALS. We report the most relevant genes implicated in ALS pathogenesis, discussing the use of different high-throughput sequencing techniques and the role of epigenomic modifications. Furthermore, we present transcriptomic studies discussing the most recent advances, from microarrays to bulk and single-cell RNA sequencing. Finally, we discuss the use of microRNAs as potential biomarkers and promising tools for molecular intervention. The integration of data from multiple omic approaches may provide new insights into pathogenic pathways in ALS by shedding light on diagnostic and prognostic biomarkers, helping to stratify patients into clinically relevant subgroups, revealing novel therapeutic targets and supporting the development of new effective therapies.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simone Scarcella
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Gianluca Costamagna
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Linda Ottoboni
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Federico Verde
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Nicola Ticozzi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| | - Delia Gagliardi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
18
|
Chang C, Zhao Q, Liu P, Yuan Y, Liu Z, Hu Y, Li W, Hou X, Tang X, Jiao B, Guo J, Shen L, Jiang H, Tang B, Zhang X, Wang J. ALS-plus related clinical and genetic study from China. Neurol Sci 2023; 44:3557-3566. [PMID: 37204564 DOI: 10.1007/s10072-023-06843-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/07/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder. An increasing number of researchers have found extra motor features in ALS, which are also called ALS-plus syndromes. Besides, a great majority of ALS patients also have cognitive impairment. However, clinical surveys of the frequency and genetic background of ALS-plus syndromes are rare, especially in China. METHODS We investigated a large cohort of 1015 patients with ALS, classifying them into six groups according to different extramotor symptoms and documenting their clinical manifestations. Meanwhile, based on their cognitive function, we divided these patients into two groups and compared demographic characteristics. Genetic screening for rare damage variants (RDVs) was also performed on 847 patients. RESULTS As a result, 16.75% of patients were identified with ALS-plus syndrome, and 49.5% of patients suffered cognitive impairment. ALS-plus group had lower ALSFRS-R scores, longer diagnostic delay time, and longer survival times, compared to ALS pure group. RDVs occurred less frequently in ALS-plus patients than in ALS-pure patients (P = 0.042) but showed no difference between ALS-cognitive impairment patients and ALS-cognitive normal patients. Besides, ALS-cognitive impairment group tends to harbour more ALS-plus symptoms than ALS-cognitive normal group (P = 0.001). CONCLUSION In summary, ALS-plus patients in China are not rare and show multiple differences from ALS-pure patients in clinical and genetic features. Besides, ALS-cognitive impairment group tends to harbour more ALS-plus syndrome than ALS-cognitive normal group. Our observations correspond with the theory that ALS involves several diseases with different mechanisms and provide clinical validation.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
- Health Management Center, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Qianqian Zhao
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Pan Liu
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Yanchun Yuan
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Zhen Liu
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Yiting Hu
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Wanzhen Li
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Xiaorong Hou
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Xuxiong Tang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Xuewei Zhang
- Health Management Center, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, People's Republic of China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, People's Republic of China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, People's Republic of China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
19
|
Liu XD, Jin T, Tao Y, Zhang M, Zheng HL, Liu QQ, Yang KH, Wei RN, Li SY, Huang Y, Xue ZY, Hao LY, Wang QH, Yang L, Lin FQ, Shen W, Tao YX, Wang HJ, Cao JL, Pan ZQ. DHX9/DNA-tandem repeat-dependent downregulation of ciRNA-Fmn1 in the dorsal horn is required for neuropathic pain. Acta Pharmacol Sin 2023; 44:1748-1767. [PMID: 37095197 PMCID: PMC10462628 DOI: 10.1038/s41401-023-01082-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/22/2023] [Indexed: 04/26/2023]
Abstract
Circular RNAs (ciRNAs) are emerging as new players in the regulation of gene expression. However, how ciRNAs are involved in neuropathic pain is poorly understood. Here, we identify the nervous-tissue-specific ciRNA-Fmn1 and report that changes in ciRNA-Fmn1 expression in spinal cord dorsal horn neurons play a key role in neuropathic pain after nerve injury. ciRNA-Fmn1 was significantly downregulated in ipsilateral dorsal horn neurons after peripheral nerve injury, at least in part because of a decrease in DNA helicase 9 (DHX9), which regulates production of ciRNA-Fmn1 by binding to DNA-tandem repeats. Blocking ciRNA-Fmn1 downregulation reversed nerve-injury-induced reductions in both the binding of ciRNA-Fmn1 to the ubiquitin ligase UBR5 and the level of ubiquitination of albumin (ALB), thereby abrogating the nerve-injury-induced increase of ALB expression in the dorsal horn and attenuating the associated pain hypersensitivities. Conversely, mimicking downregulation of ciRNA-Fmn1 in naïve mice reduced the UBR5-controlled ubiquitination of ALB, leading to increased expression of ALB in the dorsal horn and induction of neuropathic-pain-like behaviors in naïve mice. Thus, ciRNA-Fmn1 downregulation caused by changes in binding of DHX9 to DNA-tandem repeats contributes to the genesis of neuropathic pain by negatively modulating UBR5-controlled ALB expression in the dorsal horn.
Collapse
Affiliation(s)
- Xiao-Dan Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Tong Jin
- Department of Pain, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Yang Tao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ming Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hong-Li Zheng
- Department of Pain, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Qiao-Qiao Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ke-Hui Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ru-Na Wei
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Si-Yuan Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yue Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhou-Ya Xue
- Department of Anesthesiology, Yancheng Affiliated Hospital of Xuzhou Medical University, Yancheng, 224001, China
| | - Ling-Yun Hao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Qi-Hui Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Li Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Fu-Qing Lin
- Department of Pain, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Wen Shen
- Department of Pain, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Hong-Jun Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Zhi-Qiang Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
20
|
Bonato G, Mozzetta S, Bussè C, Cecchin D, Cagnin A. Frontotemporal dementia phenotype in late-onset Huntington disease without chorea. Neurol Sci 2023; 44:3327-3329. [PMID: 37140830 DOI: 10.1007/s10072-023-06823-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023]
Affiliation(s)
- Giulia Bonato
- Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
| | - Stefano Mozzetta
- Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Cinzia Bussè
- Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Diego Cecchin
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
- Nuclear Medicine Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Annachiara Cagnin
- Department of Neuroscience, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.
| |
Collapse
|
21
|
Akçimen F, Lopez ER, Landers JE, Nath A, Chiò A, Chia R, Traynor BJ. Amyotrophic lateral sclerosis: translating genetic discoveries into therapies. Nat Rev Genet 2023; 24:642-658. [PMID: 37024676 PMCID: PMC10611979 DOI: 10.1038/s41576-023-00592-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2023] [Indexed: 04/08/2023]
Abstract
Recent advances in sequencing technologies and collaborative efforts have led to substantial progress in identifying the genetic causes of amyotrophic lateral sclerosis (ALS). This momentum has, in turn, fostered the development of putative molecular therapies. In this Review, we outline the current genetic knowledge, emphasizing recent discoveries and emerging concepts such as the implication of distinct types of mutation, variability in mutated genes in diverse genetic ancestries and gene-environment interactions. We also propose a high-level model to synthesize the interdependent effects of genetics, environmental and lifestyle factors, and ageing into a unified theory of ALS. Furthermore, we summarize the current status of therapies developed on the basis of genetic knowledge established for ALS over the past 30 years, and we discuss how developing treatments for ALS will advance our understanding of targeting other neurological diseases.
Collapse
Affiliation(s)
- Fulya Akçimen
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Elia R Lopez
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Adriano Chiò
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy
- Azienda Ospedaliero Universitaria Citta' della Salute e della Scienza, Turin, Italy
| | - Ruth Chia
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Bryan J Traynor
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA.
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA.
| |
Collapse
|
22
|
Clarke AJ, Manser D, Fleischer R, Fulham M, Ahmed RM. Pearls & Oy-sters: Huntington Disease Presenting as Primary Progressive Aphasia: A Case of Semantics. Neurology 2023; 101:414-417. [PMID: 37202171 PMCID: PMC10501099 DOI: 10.1212/wnl.0000000000207428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/04/2023] [Indexed: 05/20/2023] Open
Abstract
We present a case of semantic variant primary progressive aphasia as the presenting feature in a patient with Huntington disease (HD). The patient initially developed progressive language impairment including impaired naming and object knowledge and single-word comprehension and then developed chorea and behavioral changes. An MRI of the brain showed left anterior temporal lobe and hippocampal atrophy. A neurologic FDG PET/CT showed reduced metabolism in the head of the left caudate nucleus. Huntingtin gene testing revealed an expansion of 39 CAG repeats in 1 allele. This case outlines the substantial overlap between the clinical presentation of HD and frontotemporal lobar degeneration syndromes and provides commentary on the investigation of these neurodegenerative diseases.
Collapse
Affiliation(s)
- Antonia J Clarke
- From the Faculty of Medicine and Health (A.J.C., R.M.A.), University of Sydney; Department of Neurology (A.J.C., D.M., M.F., R.M.A.), Genetics Department (R.F.), and Department of Molecular Imaging (M.F.), Royal Prince Alfred Hospital; and Faculty of Engineering and Computer Science (M.F.), University of Sydney, Australia.
| | - David Manser
- From the Faculty of Medicine and Health (A.J.C., R.M.A.), University of Sydney; Department of Neurology (A.J.C., D.M., M.F., R.M.A.), Genetics Department (R.F.), and Department of Molecular Imaging (M.F.), Royal Prince Alfred Hospital; and Faculty of Engineering and Computer Science (M.F.), University of Sydney, Australia
| | - Ronald Fleischer
- From the Faculty of Medicine and Health (A.J.C., R.M.A.), University of Sydney; Department of Neurology (A.J.C., D.M., M.F., R.M.A.), Genetics Department (R.F.), and Department of Molecular Imaging (M.F.), Royal Prince Alfred Hospital; and Faculty of Engineering and Computer Science (M.F.), University of Sydney, Australia
| | - Michael Fulham
- From the Faculty of Medicine and Health (A.J.C., R.M.A.), University of Sydney; Department of Neurology (A.J.C., D.M., M.F., R.M.A.), Genetics Department (R.F.), and Department of Molecular Imaging (M.F.), Royal Prince Alfred Hospital; and Faculty of Engineering and Computer Science (M.F.), University of Sydney, Australia
| | - Rebekah M Ahmed
- From the Faculty of Medicine and Health (A.J.C., R.M.A.), University of Sydney; Department of Neurology (A.J.C., D.M., M.F., R.M.A.), Genetics Department (R.F.), and Department of Molecular Imaging (M.F.), Royal Prince Alfred Hospital; and Faculty of Engineering and Computer Science (M.F.), University of Sydney, Australia
| |
Collapse
|
23
|
Gondim FDAA, Pinto WBVDR, Chieia MAT, Correia CDC, Cunha FMB, Dourado MET, França Júnior MC, Marques Júnior W, Oliveira ASB, Rodrigues CL, Silva DJD, Dias-Tosta E. Definitions, phenomenology, diagnosis, and management of the disorders of laughter and crying in amyotrophic lateral sclerosis (ALS): Consensus from ALS and Motor Neuron Disease Scientific Department of the Brazilian Academy of Neurology. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:764-775. [PMID: 37647907 PMCID: PMC10468253 DOI: 10.1055/s-0043-1771176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The spectrum of neuropsychiatric phenomena observed in amyotrophic lateral sclerosis (ALS) is wide and not fully understood. Disorders of laughter and crying stand among the most common manifestations. The aim of this study is to report the results of an educational consensus organized by the Brazilian Academy of Neurology to evaluate the definitions, phenomenology, diagnosis, and management of the disorders of laughter and crying in ALS patients. Twelve members of the Brazilian Academy of Neurology - considered to be experts in the field - were recruited to answer 12 questions about the subject. After exchanging revisions, a first draft was prepared. A face-to-face meeting was held in Fortaleza, Brazil on 9.23.22 to discuss it. The revised version was subsequently emailed to all members of the ALS Scientific Department from the Brazilian Academy of Neurology and the final revised version submitted for publication. The prevalence of pseudobulbar affect/pathological laughter and crying (PBA/PLC) in ALS patients from 15 combined studies and 3906 patients was 27.4% (N = 1070), ranging from 11.4% to 71%. Bulbar onset is a risk factor but there are limited studies evaluating the differences in prevalence among the different motor neuron diseases subtypes, including patients with and without frontotemporal dementia. Antidepressants and a combination of dextromethorphan and quinidine (not available in Brazil) are possible therapeutic options. This group of panelists acknowledge the multiple gaps in the current literature and reinforces the need for further studies.
Collapse
Affiliation(s)
- Francisco de Assis Aquino Gondim
- Universidade Federal do Ceará, Departamento de Clínica Médica, Núcleo de Desenvolvimento e Pesquisa de Medicamentos, Fortaleza CE, Brazil
| | - Wladimir Bocca Vieira de Rezende Pinto
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia/Ebserh, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | - Marco Antônio Troccoli Chieia
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia/Ebserh, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | | | | | | | | | - Wilson Marques Júnior
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Neurociências, Ribeirão Preto SP, Brazil
| | - Acary Souza Bulle Oliveira
- Universidade Federal de São Paulo, Departamento de Neurologia e Neurocirurgia/Ebserh, Setor de Investigações nas Doenças Neuromusculares, São Paulo SP, Brazil
| | | | - Delson José da Silva
- Universidade Federal de Goiás, Hospital das Clínicas, Unidade de Neurologia e Neurocirurgia/Ebserh, Goiânia GO, Brazil
| | - Elza Dias-Tosta
- Comissão de Ética da Academia Brasileira de Neurologia, São Paulo SP, Brazil
| |
Collapse
|
24
|
Agin-Liebes J, Hickman RA, Vonsattel JP, Faust PL, Flowers X, Sosunova IU, Ntiri J, Mayeux R, Surface M, Marder K, Fahn S, Przedborski S, Alcalay RN. Patterns of TDP-43 Deposition in Brains with LRRK2 G2019S Mutations. Mov Disord 2023; 38:1541-1545. [PMID: 37218402 PMCID: PMC10524857 DOI: 10.1002/mds.29449] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 03/09/2023] [Accepted: 05/01/2023] [Indexed: 05/24/2023] Open
Abstract
OBJECTIVE To assess for TDP-43 deposits in brains with and without a LRRK2 G2019S mutation. BACKGROUND LRRK2 G2019S mutations have been associated with parkinsonism and a wide range of pathological findings. There are no systematic studies examining the frequency and extent of TDP-43 deposits in neuropathological samples from LRRK2 G2019S carriers. METHODS Twelve brains with LRRK2 G2019S mutations were available for study from the New York Brain Bank at Columbia University; 11 of them had samples available for TDP-43 immunostaining. Clinical, demographic, and pathological data are reported for 11 brains with a LRRK2 G2019S mutation and compared to 11 brains without GBA1 or LRRK2 G2019S mutations with a pathologic diagnosis of Parkinson's disease (PD) or diffuse Lewy body disease. They were frequency matched by age, gender, parkinsonism age of onset, and disease duration. RESULTS TDP-43 aggregates were present in 73% (n = 8) of brains with a LRRK2 mutation and 18% (n = 2) of brains without a LRRK2 mutation (P = 0.03). In one brain with a LRRK2 mutation, TDP-43 proteinopathy was the primary neuropathological change. CONCLUSIONS Extranuclear TDP-43 aggregates are observed with greater frequency in LRRK2 G2019S autopsies compared to PD cases without a LRRK2 G2019S mutation. The association between LRRK2 and TDP-43 should be further explored. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Julian Agin-Liebes
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Richard A. Hickman
- Department of Defense/Uniformed Services University Brain Tissue Repository, Uniformed Services University, Bethesda, MD, 20817, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jean Paul Vonsattel
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, 630 W 168th Street, New York, NY, 10032, USA
| | - Phyllis L. Faust
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, 630 W 168th Street, New York, NY, 10032, USA
| | - Xena Flowers
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, 630 W 168th Street, New York, NY, 10032, USA
| | | | - Joel Ntiri
- Columbia College, 1130 Amsterdam Ave, New York, NY 10027, USA
| | - Richard Mayeux
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Matthew Surface
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
- The Michael J. Fox Foundation for Parkinson’s Research, New York, New York, USA
| | - Karen Marder
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Stanley Fahn
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Serge Przedborski
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, 630 W 168th Street, New York, NY, 10032, USA
- Department of Neuroscience Columbia University, 630 W 168th Street, New York, NY, 10032, USA
| | - Roy N. Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
- Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
25
|
Kaivola K, Chia R, Ding J, Rasheed M, Fujita M, Menon V, Walton RL, Collins RL, Billingsley K, Brand H, Talkowski M, Zhao X, Dewan R, Stark A, Ray A, Solaiman S, Alvarez Jerez P, Malik L, Dawson TM, Rosenthal LS, Albert MS, Pletnikova O, Troncoso JC, Masellis M, Keith J, Black SE, Ferrucci L, Resnick SM, Tanaka T, PROSPECT Consortium, Topol E, Torkamani A, Tienari P, Foroud TM, Ghetti B, Landers JE, Ryten M, Morris HR, Hardy JA, Mazzini L, D'Alfonso S, Moglia C, Calvo A, Serrano GE, Beach TG, Ferman T, Graff-Radford NR, Boeve BF, Wszolek ZK, Dickson DW, Chiò A, Bennett DA, De Jager PL, Ross OA, Dalgard CL, Gibbs JR, Traynor BJ, Scholz SW. Genome-wide structural variant analysis identifies risk loci for non-Alzheimer's dementias. CELL GENOMICS 2023; 3:100316. [PMID: 37388914 PMCID: PMC10300553 DOI: 10.1016/j.xgen.2023.100316] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/21/2023] [Accepted: 04/06/2023] [Indexed: 07/01/2023]
Abstract
We characterized the role of structural variants, a largely unexplored type of genetic variation, in two non-Alzheimer's dementias, namely Lewy body dementia (LBD) and frontotemporal dementia (FTD)/amyotrophic lateral sclerosis (ALS). To do this, we applied an advanced structural variant calling pipeline (GATK-SV) to short-read whole-genome sequence data from 5,213 European-ancestry cases and 4,132 controls. We discovered, replicated, and validated a deletion in TPCN1 as a novel risk locus for LBD and detected the known structural variants at the C9orf72 and MAPT loci as associated with FTD/ALS. We also identified rare pathogenic structural variants in both LBD and FTD/ALS. Finally, we assembled a catalog of structural variants that can be mined for new insights into the pathogenesis of these understudied forms of dementia.
Collapse
Affiliation(s)
- Karri Kaivola
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ruth Chia
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Memoona Rasheed
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Masashi Fujita
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY, USA
| | - Ronald L. Walton
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, USA
| | - Ryan L. Collins
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.), Cambridge, MA, USA
- Division of Medical Sciences and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kimberley Billingsley
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Centre for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, USA
| | - Harrison Brand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.), Cambridge, MA, USA
- Division of Medical Sciences and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.), Cambridge, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Xuefang Zhao
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.), Cambridge, MA, USA
| | - Ramita Dewan
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Ali Stark
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Anindita Ray
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Sultana Solaiman
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Pilar Alvarez Jerez
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Centre for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, USA
| | - Laksh Malik
- Centre for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, USA
| | - Ted M. Dawson
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Neuroregeneration and Stem Cell Programs, Institute of Cell Engineering, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Liana S. Rosenthal
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Olga Pletnikova
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY, USA
- Department of Pathology (Neuropathology), Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Juan C. Troncoso
- Department of Pathology (Neuropathology), Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Mario Masellis
- Cognitive & Movement Disorders Clinic, Sunnybrook Health Sciences Centre, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, Canada
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, Canada
| | - Julia Keith
- Department of Anatomical Pathology, Sunnybrook Health Sciences Centre, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
| | - Sandra E. Black
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, Canada
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
| | - Luigi Ferrucci
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Toshiko Tanaka
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD, USA
| | - PROSPECT Consortium
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY, USA
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.), Cambridge, MA, USA
- Division of Medical Sciences and Department of Medicine, Harvard Medical School, Boston, MA, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Centre for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Neuroregeneration and Stem Cell Programs, Institute of Cell Engineering, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY, USA
- Department of Pathology (Neuropathology), Johns Hopkins University Medical Center, Baltimore, MD, USA
- Cognitive & Movement Disorders Clinic, Sunnybrook Health Sciences Centre, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, Canada
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, Canada
- Department of Anatomical Pathology, Sunnybrook Health Sciences Centre, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON, Canada
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD, USA
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA, USA
- Translational Immunology, Research Programs Unit, University of Helsinki, Helsinki, Finland
- Department of Neurology, Helsinki University Hospital, Helsinki, Finland
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Genetics and Genomic Medicine Research & Teaching, UCL GOS Institute of Child Health, University College London, London, UK
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- UK Dementia Research Institute, Department of Neurogenerative Disease and Reta Lila Weston Institute, London, UK
- Institute of Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Maggiore della Carita University Hospital, Novara, Italy
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero Universitaria Città, della Salute e della Scienza, Corso Bramante, 88, Turin, Italy
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
- Department of Psychiatry and Psychology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, USA
- Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
- Institute of Cognitive Sciences and Technologies, C.N.R., Via S. Martino della Battaglia, 44, Rome, Italy
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- RNA Therapeutics Laboratory, Therapeutics Development Branch, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Eric Topol
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA, USA
| | - Ali Torkamani
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA, USA
| | - Pentti Tienari
- Translational Immunology, Research Programs Unit, University of Helsinki, Helsinki, Finland
- Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Tatiana M. Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John E. Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Mina Ryten
- Department of Genetics and Genomic Medicine Research & Teaching, UCL GOS Institute of Child Health, University College London, London, UK
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - John A. Hardy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- UK Dementia Research Institute, Department of Neurogenerative Disease and Reta Lila Weston Institute, London, UK
- Institute of Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | | | - Sandra D'Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Cristina Moglia
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero Universitaria Città, della Salute e della Scienza, Corso Bramante, 88, Turin, Italy
| | - Andrea Calvo
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero Universitaria Città, della Salute e della Scienza, Corso Bramante, 88, Turin, Italy
| | - Geidy E. Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Tanis Ferman
- Department of Psychiatry and Psychology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, USA
| | | | | | - Zbigniew K. Wszolek
- Institute of Cognitive Sciences and Technologies, C.N.R., Via S. Martino della Battaglia, 44, Rome, Italy
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, USA
| | - Adriano Chiò
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero Universitaria Città, della Salute e della Scienza, Corso Bramante, 88, Turin, Italy
- Institute of Cognitive Sciences and Technologies, C.N.R., Via S. Martino della Battaglia, 44, Rome, Italy
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY, USA
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, USA
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - J. Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Bryan J. Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
- RNA Therapeutics Laboratory, Therapeutics Development Branch, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Sonja W. Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| |
Collapse
|
26
|
Bai D, Zhu L, Jia Q, Duan X, Chen L, Wang X, Hou J, Jiang G, Yang S, Li S, Li XJ, Yin P. Loss of TDP-43 promotes somatic CAG repeat expansion in Huntington's disease knock-in mice. Prog Neurobiol 2023:102484. [PMID: 37315918 DOI: 10.1016/j.pneurobio.2023.102484] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 04/26/2023] [Accepted: 06/09/2023] [Indexed: 06/16/2023]
Abstract
TAR binding protein 43 (TDP-43) is normally present in the nucleus but mislocalized in the cytoplasm in a number of neurodegenerative diseases including Huntington's disease (HD). The nuclear loss of TDP-43 impairs gene transcription and regulation. However, it remains to be investigated whether loss of TDP-43 influences trinucleotide CAG repeat expansion in the HD gene, a genetic cause for HD. Here we report that CRISPR/Cas9 mediated-knock down of endogenous TDP-43 in the striatum of HD knock-in mice promoted CAG repeat expansion, accompanied by the increased expression of the DNA mismatch repair genes, Msh3 and Mlh1, which have been reported to increase trinucleotide repeat instability. Furthermore, suppressing Msh3 and Mlh1 by CRISPR/Cas9 targeting diminished the CAG repeat expansion. These findings suggest that nuclear TDP-43 deficiency may dysregulate the expression of DNA mismatch repair genes, leading to CAG repeat expansion and contributing to the pathogenesis of CAG repeat diseases. DATA AVAILABILITY: The key data supporting the findings of this study are presented within the article and the Supplemental Information. The RNA sequencing reported in this paper can be found at https://doi.org/10.6084/m9.figshare.22639429.
Collapse
Affiliation(s)
- Dazhang Bai
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632; Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of neurological diseases, North Sichuan Medical College, Nanchong, China, 637000
| | - Longhong Zhu
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Qingqing Jia
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Xuezhi Duan
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Laiqiang Chen
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Xiang Wang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Junqi Hou
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Guohui Jiang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632; Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of neurological diseases, North Sichuan Medical College, Nanchong, China, 637000
| | - Su Yang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632.
| | - Peng Yin
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632.
| |
Collapse
|
27
|
Santarelli S, Londero C, Soldano A, Candelaresi C, Todeschini L, Vernizzi L, Bellosta P. Drosophila melanogaster as a model to study autophagy in neurodegenerative diseases induced by proteinopathies. Front Neurosci 2023; 17:1082047. [PMID: 37274187 PMCID: PMC10232775 DOI: 10.3389/fnins.2023.1082047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/14/2023] [Indexed: 06/06/2023] Open
Abstract
Proteinopathies are a large group of neurodegenerative diseases caused by both genetic and sporadic mutations in particular genes which can lead to alterations of the protein structure and to the formation of aggregates, especially toxic for neurons. Autophagy is a key mechanism for clearing those aggregates and its function has been strongly associated with the ubiquitin-proteasome system (UPS), hence mutations in both pathways have been associated with the onset of neurodegenerative diseases, particularly those induced by protein misfolding and accumulation of aggregates. Many crucial discoveries regarding the molecular and cellular events underlying the role of autophagy in these diseases have come from studies using Drosophila models. Indeed, despite the physiological and morphological differences between the fly and the human brain, most of the biochemical and molecular aspects regulating protein homeostasis, including autophagy, are conserved between the two species.In this review, we will provide an overview of the most common neurodegenerative proteinopathies, which include PolyQ diseases (Huntington's disease, Spinocerebellar ataxia 1, 2, and 3), Amyotrophic Lateral Sclerosis (C9orf72, SOD1, TDP-43, FUS), Alzheimer's disease (APP, Tau) Parkinson's disease (a-syn, parkin and PINK1, LRRK2) and prion diseases, highlighting the studies using Drosophila that have contributed to understanding the conserved mechanisms and elucidating the role of autophagy in these diseases.
Collapse
Affiliation(s)
- Stefania Santarelli
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Chiara Londero
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Alessia Soldano
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Carlotta Candelaresi
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Leonardo Todeschini
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Luisa Vernizzi
- Institute of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
- Department of Medicine, NYU Langone Medical Center, New York, NY, United States
| |
Collapse
|
28
|
Henden L, Fearnley LG, Grima N, McCann EP, Dobson-Stone C, Fitzpatrick L, Friend K, Hobson L, Chan Moi Fat S, Rowe DB, D'Silva S, Kwok JB, Halliday GM, Kiernan MC, Mazumder S, Timmins HC, Zoing M, Pamphlett R, Adams L, Bahlo M, Blair IP, Williams KL. Short tandem repeat expansions in sporadic amyotrophic lateral sclerosis and frontotemporal dementia. SCIENCE ADVANCES 2023; 9:eade2044. [PMID: 37146135 PMCID: PMC10162670 DOI: 10.1126/sciadv.ade2044] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Pathogenic short tandem repeat (STR) expansions cause over 20 neurodegenerative diseases. To determine the contribution of STRs in sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), we used ExpansionHunter, REviewer, and polymerase chain reaction validation to assess 21 neurodegenerative disease-associated STRs in whole-genome sequencing data from 608 patients with sporadic ALS, 68 patients with sporadic FTD, and 4703 matched controls. We also propose a data-derived outlier detection method for defining allele thresholds in rare STRs. Excluding C9orf72 repeat expansions, 17.6% of clinically diagnosed ALS and FTD cases had at least one expanded STR allele reported to be pathogenic or intermediate for another neurodegenerative disease. We identified and validated 162 disease-relevant STR expansions in C9orf72 (ALS/FTD), ATXN1 [spinal cerebellar ataxia type 1 (SCA1)], ATXN2 (SCA2), ATXN8 (SCA8), TBP (SCA17), HTT (Huntington's disease), DMPK [myotonic dystrophy type 1 (DM1)], CNBP (DM2), and FMR1 (fragile-X disorders). Our findings suggest clinical and pathological pleiotropy of neurodegenerative disease genes and highlight their importance in ALS and FTD.
Collapse
Affiliation(s)
- Lyndal Henden
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Liam G Fearnley
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Natalie Grima
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Emily P McCann
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Carol Dobson-Stone
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Lauren Fitzpatrick
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Kathryn Friend
- SA Pathology, Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Lynne Hobson
- SA Pathology, Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Sandrine Chan Moi Fat
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Dominic B Rowe
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Susan D'Silva
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - John B Kwok
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Srestha Mazumder
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Hannah C Timmins
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Margaret Zoing
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Roger Pamphlett
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- Discipline of Pathology, The University of Sydney, Sydney, NSW 2050, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Lorel Adams
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Ian P Blair
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Kelly L Williams
- Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
29
|
Hickman RA, Traynor BJ, Marder KS, Vonsattel JP. Reply: More than a co-incidence? Exploring the increased frequency of amyotrophic lateral sclerosis in Huntington disease. Acta Neuropathol 2023; 145:259-261. [PMID: 36542109 PMCID: PMC10132522 DOI: 10.1007/s00401-022-02532-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Richard A Hickman
- Department of Defense/Uniformed Services University Brain Tissue Repository, Uniformed Services University, Bethesda, MD, 20817, USA.
- Human Oncogenesis and Pathogenesis Program, Sloan Kettering Institute, New York, NY, 10065, USA.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, 20892, USA
| | - Karen S Marder
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jean-Paul Vonsattel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 710 West 168th Street, New York, NY, 10032, USA
| |
Collapse
|
30
|
Bakels HS, Feleus S, van Dis V, de Bot ST. More than a co-incidence? Comment on: Amyotrophic lateral sclerosis is over-represented in two Huntington's disease brain bank cohorts: further evidence to support genetic pleiotropy of pathogenic HTT gene expansion. Acta Neuropathol 2023; 145:257-258. [PMID: 36335527 PMCID: PMC9849160 DOI: 10.1007/s00401-022-02517-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Hannah S. Bakels
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephanie Feleus
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vera van Dis
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Susanne T. de Bot
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
31
|
Bernal‐Chico A, Tepavcevic V, Manterola A, Utrilla C, Matute C, Mato S. Endocannabinoid signaling in brain diseases: Emerging relevance of glial cells. Glia 2023; 71:103-126. [PMID: 35353392 PMCID: PMC9790551 DOI: 10.1002/glia.24172] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
The discovery of cannabinoid receptors as the primary molecular targets of psychotropic cannabinoid Δ9 -tetrahydrocannabinol (Δ9 -THC) in late 1980s paved the way for investigations on the effects of cannabis-based therapeutics in brain pathology. Ever since, a wealth of results obtained from studies on human tissue samples and animal models have highlighted a promising therapeutic potential of cannabinoids and endocannabinoids in a variety of neurological disorders. However, clinical success has been limited and major questions concerning endocannabinoid signaling need to be satisfactorily addressed, particularly with regard to their role as modulators of glial cells in neurodegenerative diseases. Indeed, recent studies have brought into the limelight diverse, often unexpected functions of astrocytes, oligodendrocytes, and microglia in brain injury and disease, thus providing scientific basis for targeting glial cells to treat brain disorders. This Review summarizes the current knowledge on the molecular and cellular hallmarks of endocannabinoid signaling in glial cells and its clinical relevance in neurodegenerative and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Ana Bernal‐Chico
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Neuroimmunology UnitBiocruces BizkaiaBarakaldoSpain
| | | | - Andrea Manterola
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Present address:
Parque Científico y Tecnológico de GuipuzkoaViralgenSan SebastianSpain
| | | | - Carlos Matute
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Susana Mato
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Neuroimmunology UnitBiocruces BizkaiaBarakaldoSpain
| |
Collapse
|
32
|
Jih KY, Lai KL, Lin KP, Liao YC, Lee YC. Reduced-penetrance Huntington's disease-causing alleles with 39 CAG trinucleotide repeats could be a genetic factor of amyotrophic lateral sclerosis. J Chin Med Assoc 2023; 86:47-51. [PMID: 36599142 DOI: 10.1097/jcma.0000000000000837] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Expanded HTT alleles with 40 or more CAG repeats were recently found to be a rare cause of frontotemporal dementia and amyotrophic lateral sclerosis (ALS) spectrum diseases. The aim of this study was to investigate the role of HTT repeat expansions in a Taiwanese cohort with ALS. METHODS We analyzed the numbers of CAG repeats in exon 1 of HTT in a cohort of 410 Taiwanese patients with ALS and 1514 control individuals by utilizing polymerase chain reaction and amplicon fragment length analysis. RESULTS Only one of the 410 ALS patients carried a reduced-penetrance HD-causing allele with 39 CAG repeats, and none had an expanded HTT CAG repeats ≥40. The patient presented with rapidly progressive bulbar-onset ALS with disease onset at the age of 64 years. He had neither chorea nor cognitive impairment. He had a family history of chorea, but no other family member manifested with ALS. None of the 1514 control individuals carried an HTT expanded allele with CAG repeats larger than 37 repeats. CONCLUSION The HTT allele with 39 CAG repeats could be a genetic factor linked to ALS susceptibility.
Collapse
Affiliation(s)
- Kang-Yang Jih
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Physiology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Kuan-Lin Lai
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Kon-Ping Lin
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Chung Lee
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
33
|
Huntingtin and Other Neurodegeneration-Associated Proteins in the Development of Intracellular Pathologies: Potential Target Search for Therapeutic Intervention. Int J Mol Sci 2022; 23:ijms232415533. [PMID: 36555175 PMCID: PMC9779313 DOI: 10.3390/ijms232415533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are currently incurable. Numerous experimental data accumulated over the past fifty years have brought us closer to understanding the molecular and cell mechanisms responsible for their development. However, these data are not enough for a complete understanding of the genesis of these diseases, nor to suggest treatment methods. It turns out that many cellular pathologies developing during neurodegeneration coincide from disease to disease. These observations give hope to finding a common intracellular target(s) and to offering a universal method of treatment. In this review, we attempt to analyze data on similar cellular disorders among neurodegenerative diseases in general, and polyglutamine neurodegenerative diseases in particular, focusing on the interaction of various proteins involved in the development of neurodegenerative diseases with various cellular organelles. The main purposes of this review are: (1) to outline the spectrum of common intracellular pathologies and to answer the question of whether it is possible to find potential universal target(s) for therapeutic intervention; (2) to identify specific intracellular pathologies and to speculate about a possible general approach for their treatment.
Collapse
|
34
|
Watabe K, Niida-Kawaguchi M, Tada M, Kato Y, Murata M, Tanji K, Wakabayashi K, Yamada M, Kakita A, Shibata N. Praja1 RING-finger E3 ubiquitin ligase is a common suppressor of neurodegenerative disease-associated protein aggregation. Neuropathology 2022; 42:488-504. [PMID: 35701899 PMCID: PMC10084124 DOI: 10.1111/neup.12840] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/15/2022]
Abstract
The formation of misfolded protein aggregates is one of the pathological hallmarks of neurodegenerative diseases. We have previously demonstrated the cytoplasmic aggregate formation of adenovirally expressed transactivation response DNA-binding protein of 43 kDa (TDP-43), the main constituent of neuronal cytoplasmic aggregates in cases of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), in cultured neuronal cells under the condition of proteasome inhibition. The TDP-43 aggregate formation was markedly suppressed by co-infection of adenoviruses expressing heat shock transcription factor 1 (HSF1), a master regulator of heat shock response, and Praja1 RING-finger E3 ubiquitin ligase (PJA1) located downstream of the HSF1 pathway. In the present study, we examined other reportedly known E3 ubiquitin ligases for TDP-43, i.e. Parkin, RNF112 and RNF220, but failed to find their suppressive effects on neuronal cytoplasmic TDP-43 aggregate formation, although they all bind to TDP-43 as verified by co-immunoprecipitation. In contrast, PJA1 also binds to adenovirally expressed wild-type and mutated fused in sarcoma, superoxide dismutase 1, α-synuclein and ataxin-3, and huntingtin polyglutamine proteins in neuronal cultures and suppressed the aggregate formation of these proteins. These results suggest that PJA1 is a common sensing factor for aggregate-prone proteins to counteract their aggregation propensity, and could be a potential therapeutic target for neurodegenerative diseases that include ALS, FTLD, Parkinson's disease and polyglutamine diseases.
Collapse
Affiliation(s)
- Kazuhiko Watabe
- Department of Medical Technology (Neuropathology), Faculty of Health Sciences, Kyorin University, Tokyo, Japan.,Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Motoko Niida-Kawaguchi
- Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan.,Department of Clinical Psychology, Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Mari Tada
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yoichiro Kato
- Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Makiko Murata
- Department of Medical Technology (Neuropathology), Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Mitsunori Yamada
- Division of Neuropathology, Department of Brain Disease Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Noriyuki Shibata
- Division of Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
35
|
Manini A, Gagliardi D, Meneri M, Antognozzi S, Del Bo R, Scaglione C, Comi GP, Corti S, Ronchi D. Analysis of
HTT
CAG
repeat expansion in Italian patients with amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2022; 9:1820-1825. [DOI: 10.1002/acn3.51673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Arianna Manini
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
| | - Delia Gagliardi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Department of Neuroscience, Neurology Unit IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico Milan Italy
| | - Megi Meneri
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
| | - Sara Antognozzi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
| | - Roberto Del Bo
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
| | - Cesa Scaglione
- IRCCS Istituto delle Scienze Neurologiche di Bologna Bologna Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Department of Neuroscience IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit Milan Italy
| | - Stefania Corti
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Department of Neuroscience, Neurology Unit IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico Milan Italy
| | - Dario Ronchi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Department of Neuroscience, Neurology Unit IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico Milan Italy
| |
Collapse
|
36
|
Feldman EL, Goutman SA, Petri S, Mazzini L, Savelieff MG, Shaw PJ, Sobue G. Amyotrophic lateral sclerosis. Lancet 2022; 400:1363-1380. [PMID: 36116464 PMCID: PMC10089700 DOI: 10.1016/s0140-6736(22)01272-7] [Citation(s) in RCA: 464] [Impact Index Per Article: 154.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/24/2022] [Accepted: 06/23/2022] [Indexed: 01/07/2023]
Abstract
Amyotrophic lateral sclerosis is a fatal CNS neurodegenerative disease. Despite intensive research, current management of amyotrophic lateral sclerosis remains suboptimal from diagnosis to prognosis. Recognition of the phenotypic heterogeneity of amyotrophic lateral sclerosis, global CNS dysfunction, genetic architecture, and development of novel diagnostic criteria is clarifying the spectrum of clinical presentation and facilitating diagnosis. Insights into the pathophysiology of amyotrophic lateral sclerosis, identification of disease biomarkers and modifiable risks, along with new predictive models, scales, and scoring systems, and a clinical trial pipeline of mechanism-based therapies, are changing the prognostic landscape. Although most recent advances have yet to translate into patient benefit, the idea of amyotrophic lateral sclerosis as a complex syndrome is already having tangible effects in the clinic. This Seminar will outline these insights and discuss the status of the management of amyotrophic lateral sclerosis for the general neurologist, along with future prospects that could improve care and outcomes for patients with amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Eva L Feldman
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Stephen A Goutman
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Letizia Mazzini
- ALS Centre, Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy; Department of Neurology, University of Piemonte Orientale, Novara, Italy
| | - Masha G Savelieff
- Department of Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Gen Sobue
- Department of Neurology, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
37
|
Reho P, Koga S, Shah Z, Chia R, International LBD Genomics Consortium, The American Genome Center, Rademakers R, Dalgard CL, Boeve BF, Beach TG, Dickson DW, Ross OA, Scholz SW. GRN Mutations Are Associated with Lewy Body Dementia. Mov Disord 2022; 37:1943-1948. [PMID: 35810449 PMCID: PMC9474656 DOI: 10.1002/mds.29144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/30/2022] [Accepted: 06/13/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Loss-of-function mutations in GRN are a cause of familial frontotemporal dementia, and common variants within the gene have been associated with an increased risk of developing Alzheimer's disease and Parkinson's disease. Although TDP-43-positive inclusions are characteristic of GRN-related neurodegeneration, Lewy body copathology has also been observed in many GRN mutation carriers. OBJECTIVE The objective of this study was to assess a Lewy body dementia (LBD) case-control cohort for pathogenic variants in GRN and to test whether there is an enrichment of damaging mutations among patients with LBD. METHODS We analyzed whole-genome sequencing data generated for 2591 European-ancestry LBD cases and 4032 neurologically healthy control subjects to identify disease-causing mutations in GRN. RESULTS We identified six heterozygous exonic GRN mutations in seven study participants (cases: n = 6; control subjects: n = 1). Each variant was predicted to be pathogenic or likely pathogenic. We found significant enrichment of GRN loss-of-function mutations in patients with LBD compared with control subjects (Optimized Sequence Kernel Association Test P = 0.0162). Immunohistochemistry in three definite LBD cases demonstrated Lewy body pathology and TDP-43-positive neuronal inclusions. CONCLUSIONS Our findings suggest that deleterious GRN mutations are a rare cause of familial LBD. © 2022 International Parkinson Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Paolo Reho
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Zalak Shah
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ruth Chia
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | | | | | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, USA
| | - Sonja W. Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| |
Collapse
|
38
|
Cao MC, Scotter EL. Novel and known transcriptional targets of ALS/FTD protein TDP-43: Meta-analysis and interactive graphical database. Dis Model Mech 2022; 15:276263. [PMID: 35946434 PMCID: PMC9509890 DOI: 10.1242/dmm.049418] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
TDP-43 proteinopathy is the major pathology in amyotrophic lateral sclerosis (ALS) and tau-negative frontotemporal dementia (FTD). Mounting evidence implicates loss of normal TDP-43 RNA processing function as a key pathomechanism. However, the RNA targets of TDP-43 differ by report, and have never been formally collated or compared between models and disease, hampering understanding of TDP-43 function. Here, we conducted re-analysis and meta-analysis of publicly available RNA-sequencing datasets from six TDP-43-knockdown models, and TDP-43-immunonegative neuronal nuclei from ALS/ FTD brain, to identify differentially expressed genes (DEGs) and exon usage (DEU) events. There was little overlap in DEGs between knockdown models, but PFKP, STMN2, CFP, KIAA1324 and TRHDE were common targets and were also differentially expressed in TDP-43-immunonegative neurons. DEG enrichment analysis revealed diverse biological pathways including immune and synaptic functions. Common DEU events in human datasets included well-known targets POLDIP3 and STMN2, and novel targets EXD3, MMAB, DLG5 and GOSR2. Our interactive database https://phpstack-449938-2576646.cloudwaysapps.com/ allows further exploration of TDP-43 DEG and DEU targets. Together, these data identify TDP-43 targets that can be exploited therapeutically or to validate loss-of-function processes.
Collapse
Affiliation(s)
- Maize C Cao
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand. 3A Symonds Street, Auckland 1010, New Zealand
| | - Emma L Scotter
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand. 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
39
|
Grassano M, Calvo A, Moglia C, Sbaiz L, Brunetti M, Barberis M, Casale F, Manera U, Vasta R, Canosa A, D'Alfonso S, Corrado L, Mazzini L, Dalgard C, Karra R, Chia R, Traynor B, Chiò A. Systematic evaluation of genetic mutations in ALS: a population-based study. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2022-328931. [PMID: 35896380 PMCID: PMC9606529 DOI: 10.1136/jnnp-2022-328931] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/22/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND A genetic diagnosis in Amyotrophic Lateral Sclerosis (ALS) can inform genetic counselling, prognosis and, in the light of incoming gene-targeted therapy, management. However, conventional genetic testing strategies are often costly and time-consuming. OBJECTIVE To evaluate the diagnostic yield and advantages of whole-genome sequencing (WGS) as a standard diagnostic genetic test for ALS. METHODS In this population-based cohort study, 1043 ALS patients from the Piemonte and Valle d'Aosta Register for ALS and 755 healthy individuals were screened by WGS for variants in 42 ALS-related genes and for repeated-expansions in C9orf72 and ATXN2. RESULTS A total of 279 ALS cases (26.9%) received a genetic diagnosis, namely 75.2% of patients with a family history of ALS and 21.5% of sporadic cases. The mutation rate among early-onset ALS patients was 43.9%, compared with 19.7% of late-onset patients. An additional 14.6% of the cohort carried a genetic factor that worsen prognosis. CONCLUSIONS Our results suggest that, because of its high diagnostic yield and increasingly competitive costs, along with the possibility of retrospectively reassessing newly described genes, WGS should be considered as standard genetic testing for all ALS patients. Additionally, our results provide a detailed picture of the genetic basis of ALS in the general population.
Collapse
Affiliation(s)
- Maurizio Grassano
- Department of Neuroscience, University of Turin, Torino, Italy
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, Porter Neuroscience Research Center, National Institute on Aging, Bethesda, Maryland, USA
| | - Andrea Calvo
- Department of Neuroscience, University of Turin, Torino, Italy
- S.C. Neurologia 1U, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
| | - Cristina Moglia
- Department of Neuroscience, University of Turin, Torino, Italy
- S.C. Neurologia 1U, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
| | - Luca Sbaiz
- Laboratory of Genetics, Department of Clinical Pathology, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
| | - Maura Brunetti
- Laboratory of Genetics, Department of Clinical Pathology, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
| | - Marco Barberis
- Laboratory of Genetics, Department of Clinical Pathology, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
| | - Federico Casale
- Department of Neuroscience, University of Turin, Torino, Italy
| | - Umberto Manera
- Department of Neuroscience, University of Turin, Torino, Italy
- S.C. Neurologia 1U, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
| | - Rosario Vasta
- Department of Neuroscience, University of Turin, Torino, Italy
| | - Antonio Canosa
- Department of Neuroscience, University of Turin, Torino, Italy
- S.C. Neurologia 1U, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, University of Eastern Piedmont Amedeo Avogadro School of Medicine, Novara, Piemonte, Italy
| | - Lucia Corrado
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, University of Eastern Piedmont Amedeo Avogadro School of Medicine, Novara, Piemonte, Italy
| | - Letizia Mazzini
- Department of Neurology, University Hospital Maggiore della Carità, Novara, Italy
| | - Clifton Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Ramita Karra
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, Porter Neuroscience Research Center, National Institute on Aging, Bethesda, Maryland, USA
| | - Ruth Chia
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, Porter Neuroscience Research Center, National Institute on Aging, Bethesda, Maryland, USA
| | - Bryan Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, Porter Neuroscience Research Center, National Institute on Aging, Bethesda, Maryland, USA
- Department of Neurology and Neurosurgery, Johns Hopkins University, Baltimore, Maryland, USA
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London, UK
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
- ASO Rapid Development Laboratory, Therapeutics Development Branch, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Adriano Chiò
- Department of Neuroscience, University of Turin, Torino, Italy
- S.C. Neurologia 1U, Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Torino, Italy
- Institute of Cognitive Sciences and Technologies, National Council of Research, Rome, Italy
| |
Collapse
|
40
|
Pérez‐Oliveira S, Álvarez I, Rosas I, Menendez‐González M, Blázquez‐Estrada M, Aguilar M, Corte D, Buongiorno M, Molina‐Porcel L, Aldecoa I, Martí MJ, Sánchez‐Juan P, Infante J, González‐Aramburu I, García‐González P, Rosende‐Roca M, Boada M, Ruiz A, Periñán MT, Macías‐García D, Muñoz‐Delgado L, Gómez‐Garre P, Mir P, Clarimón J, Lleo A, Alcolea D, De la Casa‐Fages B, Duarte I, Álvarez V, Pastor P. Intermediate and Expanded
HTT
Alleles and the Risk for α‐Synucleinopathies. Mov Disord 2022; 37:1841-1849. [DOI: 10.1002/mds.29153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022] Open
Affiliation(s)
| | - Ignacio Álvarez
- Movement Disorders Unit, Department of Neurology University Hospital Mútua de Terrassa and Fundació Docència i Recerca Mútua de Terrassa Terrassa, Barcelona Spain
| | - Irene Rosas
- Laboratorio de Genética Hospital Universitario Central de Asturias Oviedo Spain
| | - Manuel Menendez‐González
- Department of Neurology Hospital Universitario Central de Asturias Oviedo Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Marta Blázquez‐Estrada
- Department of Neurology Hospital Universitario Central de Asturias Oviedo Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Miquel Aguilar
- Movement Disorders Unit, Department of Neurology University Hospital Mútua de Terrassa and Fundació Docència i Recerca Mútua de Terrassa Terrassa, Barcelona Spain
| | - Daniela Corte
- Biobank of Principado de Asturias Hospital Universitario Central de Asturias (HUCA) Oviedo Spain
| | - Mariateresa Buongiorno
- Movement Disorders Unit, Department of Neurology University Hospital Mútua de Terrassa and Fundació Docència i Recerca Mútua de Terrassa Terrassa, Barcelona Spain
| | - Laura Molina‐Porcel
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department Hospital Clínic i Provincial de Barcelona and Institut d'Investigacions Biomèdiques August Pi I Sunyer Barcelona Spain
- Neurological Tissue Bank of the Biobank‐Hospital Clinic‐IDIBAPS Barcelona Spain
| | - Iban Aldecoa
- Neurological Tissue Bank of the Biobank‐Hospital Clinic‐IDIBAPS Barcelona Spain
- Pathology Department, Biomedical Diagnostic Center Hospital Clínic de Barcelona, University of Barcelona Barcelona Spain
| | - María J. Martí
- Parkinson's Disease and Movement Disorders Unit, Department of Neurology, Hospital Clinic of Barcelona, Spain; Institut de Neurociències, Maeztu Center, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) University of Barcelona Barcelona Spain
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
| | - Pascual Sánchez‐Juan
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Alzheimer’s Centre Reina Sofia‐CIEN Foundation‐ISCIII Madrid Spain
| | - Jon Infante
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Department of Neurology Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL) Santander Spain
| | - Isabel González‐Aramburu
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Department of Neurology Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL) Santander Spain
| | - Pablo García‐González
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Research Center and Memory clinic Fundació ACE, Institut Català de Neurociències Aplicades Universitat Internacional de Catalunya Barcelona Spain
| | - Maitée Rosende‐Roca
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Research Center and Memory clinic Fundació ACE, Institut Català de Neurociències Aplicades Universitat Internacional de Catalunya Barcelona Spain
| | - Mercè Boada
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Research Center and Memory clinic Fundació ACE, Institut Català de Neurociències Aplicades Universitat Internacional de Catalunya Barcelona Spain
| | - Agustín Ruiz
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Research Center and Memory clinic Fundació ACE, Institut Català de Neurociències Aplicades Universitat Internacional de Catalunya Barcelona Spain
| | - María Teresa Periñán
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Movement Disorders Unit, Department of Neurology and Neurophysiology Instituto de Biomedicina de Sevilla Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville Spain
| | - Daniel Macías‐García
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Movement Disorders Unit, Department of Neurology and Neurophysiology Instituto de Biomedicina de Sevilla Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville Spain
| | - Laura Muñoz‐Delgado
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Movement Disorders Unit, Department of Neurology and Neurophysiology Instituto de Biomedicina de Sevilla Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville Spain
| | - Pilar Gómez‐Garre
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Movement Disorders Unit, Department of Neurology and Neurophysiology Instituto de Biomedicina de Sevilla Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville Spain
| | - Pablo Mir
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Movement Disorders Unit, Department of Neurology and Neurophysiology Instituto de Biomedicina de Sevilla Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville Spain
- Department of Medicine, Facultad de Medicina Universidad de Sevilla Seville Spain
| | - Jordi Clarimón
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Department of Neurology, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - Alberto Lleo
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Department of Neurology, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - Daniel Alcolea
- CIBERNED Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III Madrid Spain
- Department of Neurology, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - Beatriz De la Casa‐Fages
- Movement Disorders Unit, Department of Neurology Hospital General Universitario Gregorio Marañón Madrid Spain
- Instituto Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Israel Duarte
- Laboratorio de Genética Hospital Universitario Central de Asturias Oviedo Spain
| | - Victoria Álvarez
- Laboratorio de Genética Hospital Universitario Central de Asturias Oviedo Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona Barcelona Spain
| |
Collapse
|
41
|
Dewan R, Jaunmuktane Z, Garcia‐Segura ME, Strand C, Wild E, Villar J, Dalgard CL, Tabrizi SJ, Traynor BJ, Proukakis C. CAG Somatic Instability in a Huntington Disease Expansion Carrier Presenting with a Progressive Supranuclear Palsy-like Phenotype. Mov Disord 2022; 37:1555-1557. [PMID: 35510647 PMCID: PMC9308656 DOI: 10.1002/mds.29035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/15/2022] [Accepted: 04/08/2022] [Indexed: 12/03/2022] Open
Affiliation(s)
- Ramita Dewan
- Neuromuscular Disease Research Section, Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | - Zane Jaunmuktane
- Department of Clinical and Movement NeurosciencesQueen Square Institute of Neurology, UCLLondonUnited Kingdom
- Queen Square Brain Bank, UCLLondonUnited Kingdom
| | - Monica Emili Garcia‐Segura
- Department of Clinical and Movement NeurosciencesQueen Square Institute of Neurology, UCLLondonUnited Kingdom
| | | | - Edward Wild
- Huntington's Disease Centre, Queen Square Institute of Neurology, UCLLondonUnited Kingdom
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCLLondonUnited Kingdom
| | - Joaquin Villar
- Center for Military Precision MedicineUniformed Services University of the Health SciencesBethesdaMarylandUSA
- Henry M Jackson Foundation for the Advancement of Military MedicineBethesdaMarylandUSA
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology, and GeneticsUniformed Services University of the Health SciencesBethesdaMarylandUSA
- The American Genome CenterUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Sarah J. Tabrizi
- Huntington's Disease Centre, Queen Square Institute of Neurology, UCLLondonUnited Kingdom
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCLLondonUnited Kingdom
- UK Dementia Research Institute, UCLLondonUnited Kingdom
| | - Bryan J. Traynor
- Neuromuscular Disease Research Section, Laboratory of NeurogeneticsNational Institute on AgingBethesdaMarylandUSA
| | - Christos Proukakis
- Department of Clinical and Movement NeurosciencesQueen Square Institute of Neurology, UCLLondonUnited Kingdom
| |
Collapse
|
42
|
Martí-Martínez S, Valor LM. A Glimpse of Molecular Biomarkers in Huntington's Disease. Int J Mol Sci 2022; 23:ijms23105411. [PMID: 35628221 PMCID: PMC9142992 DOI: 10.3390/ijms23105411] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder that is caused by an abnormal expansion of CAG repeats in the Huntingtin (HTT) gene. Although the main symptomatology is explained by alterations at the level of the central nervous system, predominantly affecting the basal ganglia, a peripheral component of the disease is being increasingly acknowledged. Therefore, the manifestation of the disease is complex and variable among CAG expansion carriers, introducing uncertainty in the appearance of specific signs, age of onset and severity of disease. The monogenic nature of the disorder allows a precise diagnosis, but the use of biomarkers with prognostic value is still needed to achieve clinical management of the patients in an individual manner. In addition, we need tools to evaluate the patient's response to potential therapeutic approaches. In this review, we provide a succinct summary of the most interesting molecular biomarkers that have been assessed in patients, mostly obtained from body fluids such as cerebrospinal fluid, peripheral blood and saliva.
Collapse
Affiliation(s)
- Silvia Martí-Martínez
- Servicio de Neurología, Hospital General Universitario Dr. Balmis, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain;
| | - Luis M. Valor
- Laboratorio de Apoyo a la Investigación, Hospital General Universitario Dr. Balmis, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
- Correspondence: ; Tel.: +34-965-913-988
| |
Collapse
|
43
|
Glass JD, Dewan R, Ding J, Gibbs JR, Dalgard C, Keagle PJ, Shankaracharya, García-Redondo A, Traynor BJ, Chia R, Landers JE. ATXN2 intermediate expansions in amyotrophic lateral sclerosis. Brain 2022; 145:2671-2676. [PMID: 35521889 PMCID: PMC9890463 DOI: 10.1093/brain/awac167] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/21/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Intermediate CAG (polyQ) expansions in the gene ataxin-2 (ATXN2) are now recognized as a risk factor for amyotrophic lateral sclerosis. The threshold for increased risk is not yet firmly established, with reports ranging from 27 to 31 repeats. We investigated the presence of ATXN2 polyQ expansions in 9268 DNA samples collected from people with amyotrophic lateral sclerosis, amyotrophic lateral sclerosis with frontotemporal dementia, frontotemporal dementia alone, Lewy body dementia and age matched controls. This analysis confirmed ATXN2 intermediate polyQ expansions of ≥31 as a risk factor for amyotrophic lateral sclerosis with an odds ratio of 6.31. Expansions were an even greater risk for amyotrophic lateral sclerosis with frontotemporal dementia (odds ratio 27.59) and a somewhat lesser risk for frontotemporal dementia alone (odds ratio 3.14). There was no increased risk for Lewy body dementia. In a subset of 1362 patients with amyotrophic lateral sclerosis with complete clinical data, we could not confirm previous reports of earlier onset of amyotrophic lateral sclerosis or shorter survival in 25 patients with expansions. These new data confirm ≥31 polyQ repeats in ATXN2 increase the risk for amyotrophic lateral sclerosis, and also for the first time show an even greater risk for amyotrophic lateral sclerosis with frontotemporal dementia. The lack of a more aggressive phenotype in amyotrophic lateral sclerosis patients with expansions has implications for ongoing gene-silencing trials for amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Jonathan D Glass
- Correspondence to: Jonathan D. Glass, MD Department of Neurology, Emory University 101 Woodruff Circle, Atlanta, GA 30322, USA E-mail:
| | - Ramita Dewan
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Jinhui Ding
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA,Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - J Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Clifton Dalgard
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Pamela J Keagle
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Shankaracharya
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
44
|
Goutman SA, Hardiman O, Al-Chalabi A, Chió A, Savelieff MG, Kiernan MC, Feldman EL. Recent advances in the diagnosis and prognosis of amyotrophic lateral sclerosis. Lancet Neurol 2022; 21:480-493. [PMID: 35334233 PMCID: PMC9513753 DOI: 10.1016/s1474-4422(21)00465-8] [Citation(s) in RCA: 232] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/24/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022]
Abstract
The diagnosis of amyotrophic lateral sclerosis can be challenging due to its heterogeneity in clinical presentation and overlap with other neurological disorders. Diagnosis early in the disease course can improve outcomes as timely interventions can slow disease progression. An evolving awareness of disease genotypes and phenotypes and new diagnostic criteria, such as the recent Gold Coast criteria, could expedite diagnosis. Improved prognosis, such as that achieved with the survival model from the European Network for the Cure of ALS, could inform the patient and their family about disease course and improve end-of-life planning. Novel staging and scoring systems can help monitor disease progression and might potentially serve as clinical trial outcomes. Lastly, new tools, such as fluid biomarkers, imaging modalities, and neuromuscular electrophysiological measurements, might increase diagnostic and prognostic accuracy.
Collapse
Affiliation(s)
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, and Department of Neurology, King's College London, London, UK
| | - Adriano Chió
- Rita Levi Montalcini Department of Neurosciences, University of Turin, Turin, Italy
| | | | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia; Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
45
|
Goutman SA, Hardiman O, Al-Chalabi A, Chió A, Savelieff MG, Kiernan MC, Feldman EL. Emerging insights into the complex genetics and pathophysiology of amyotrophic lateral sclerosis. Lancet Neurol 2022; 21:465-479. [PMID: 35334234 PMCID: PMC9513754 DOI: 10.1016/s1474-4422(21)00414-2] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/21/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis is a fatal neurodegenerative disease. The discovery of genes associated with amyotrophic lateral sclerosis, commencing with SOD1 in 1993, started fairly gradually. Recent advances in genetic technology have led to the rapid identification of multiple new genes associated with the disease, and to a new understanding of oligogenic and polygenic disease risk. The overlap of genes associated with amyotrophic lateral sclerosis with those of other neurodegenerative diseases is shedding light on the phenotypic spectrum of neurodegeneration, leading to a better understanding of genotype-phenotype correlations. A deepening knowledge of the genetic architecture is allowing the characterisation of the molecular steps caused by various mutations that converge on recurrent dysregulated pathways. Of crucial relevance, mutations associated with amyotrophic lateral sclerosis are amenable to novel gene-based therapeutic options, an approach in use for other neurological illnesses. Lastly, the exposome-the summation of lifetime environmental exposures-has emerged as an influential component for amyotrophic lateral sclerosis through the gene-time-environment hypothesis. Our improved understanding of all these aspects will lead to long-awaited therapies and the identification of modifiable risks factors.
Collapse
Affiliation(s)
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, and Department of Neurology, King's College London, London, UK
| | - Adriano Chió
- Rita Levi Montalcini Department of Neurosciences, University of Turin, Turin, Italy
| | | | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia; Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
46
|
Hickman RA, Faust PL, Marder K, Yamamoto A, Vonsattel JP. The distribution and density of Huntingtin inclusions across the Huntington disease neocortex: regional correlations with Huntingtin repeat expansion independent of pathologic grade. Acta Neuropathol Commun 2022; 10:55. [PMID: 35440014 PMCID: PMC9020040 DOI: 10.1186/s40478-022-01364-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Huntington disease is characterized by progressive neurodegeneration, especially of the striatum, and the presence of polyglutamine huntingtin (HTT) inclusions. Although HTT inclusions are most abundant in the neocortex, their neocortical distribution and density in relation to the extent of CAG repeat expansion in the HTT gene and striatal pathologic grade have yet to be formally established. We immunohistochemically studied 65 brains with a pathologic diagnosis of Huntington disease to investigate the cortical distributions and densities of HTT inclusions within the calcarine (BA17), precuneus (BA7), motor (BA4) and prefrontal (BA9) cortices; in 39 of these brains, a p62 immunostain was used for comparison. HTT inclusions predominate in the infragranular cortical layers (layers V-VI) and layer III, however, the densities of HTT inclusions across the human cerebral cortex are not uniform but are instead regionally contingent. The density of HTT and p62 inclusions (intranuclear and extranuclear) in layers V-VI increases caudally to rostrally (BA17 < BA7 < BA4 < BA9) with the median burden of HTT inclusions being 38-fold greater in the prefrontal cortex (BA9) than in the calcarine cortex (BA17). Conversely, intranuclear HTT inclusions prevail in the calcarine cortex irrespective of HTT CAG length. Neocortical HTT inclusion density correlates with CAG repeat expansion, but not with the neuropathologic grade of striatal degeneration (Vonsattel grade) or with the duration of clinical disease since motor onset. Extrapolation of these findings suggest that HTT inclusions are at a regionally-contingent, CAG-dependent, density during the advanced stages of HD. The distribution and density of HTT inclusions in HD therefore does not provide a measure of pathologic disease stage but rather infers the degree of pathogenic HTT expansion.
Collapse
Affiliation(s)
- Richard A. Hickman
- grid.51462.340000 0001 2171 9952Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065 USA
| | - Phyllis L. Faust
- grid.413734.60000 0000 8499 1112Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, 630 W 168th Street, New York, NY 10032 USA
| | - Karen Marder
- grid.21729.3f0000000419368729Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Ai Yamamoto
- grid.413734.60000 0000 8499 1112Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, 630 W 168th Street, New York, NY 10032 USA ,grid.21729.3f0000000419368729Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Jean-Paul Vonsattel
- grid.413734.60000 0000 8499 1112Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, 630 W 168th Street, New York, NY 10032 USA ,grid.239585.00000 0001 2285 2675Taub Institute for Research On Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, 710 West 168th Street, New York, NY 10032 USA
| |
Collapse
|
47
|
Madhav H, Jameel E, Rehan M, Hoda N. Recent advancements in chromone as a privileged scaffold towards the development of small molecules for neurodegenerative therapeutics. RSC Med Chem 2022; 13:258-279. [PMID: 35434628 PMCID: PMC8942243 DOI: 10.1039/d1md00394a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 01/27/2022] [Indexed: 02/02/2023] Open
Abstract
Neurodegenerative disorders, i.e., Alzheimer's or Parkinson's disease, involve progressive degeneration of the central nervous system, resulting in memory loss and cognitive impairment. The intensification of neurodegenerative research in recent years put some molecules into clinical trials, but still there is an urgent need to develop effective therapeutic molecules to combat these diseases. Chromone is a well-identified privileged structure for the design of well-diversified therapeutic molecules of potential pharmacological interest, particularly in the field of neurodegeneration. In this short review, we focused on the recent advancements and developments of chromones for neurodegenerative therapeutics. Different small molecules were reviewed as multi-target-directed ligands (MTDLs) with potential inhibition of AChE, BuChE, MAO-A, MAO-B, Aβ plaque formation and aggregation. Recently developed MTDLs emphasized that the chromone scaffold has the potential to develop new molecules for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hari Madhav
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia IslamiaNew Delhi110025India
| | - Ehtesham Jameel
- College of Pharmaceutical Sciences, Zhejiang UniversityHangzhouPR China
| | - Mohammad Rehan
- Max-Planck-Institute für Molekulare Physiologie, Abteilung Chemische BiologieOtto-Hahn-Straße 1144227 DortmundGermany
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia IslamiaNew Delhi110025India
| |
Collapse
|
48
|
Ain Q, Schmeer CW, Wengerodt D, Hofmann Y, Witte OW, Kretz A. Optimized Protocol for Proportionate CNS Cell Retrieval as a Versatile Platform for Cellular and Molecular Phenomapping in Aging and Neurodegeneration. Int J Mol Sci 2022; 23:ijms23063000. [PMID: 35328432 PMCID: PMC8950438 DOI: 10.3390/ijms23063000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/04/2023] Open
Abstract
Efficient purification of viable neural cells from the mature CNS has been historically challenging due to the heterogeneity of the inherent cell populations. Moreover, changes in cellular interconnections, membrane lipid and cholesterol compositions, compartment-specific biophysical properties, and intercellular space constituents demand technical adjustments for cell isolation at different stages of maturation and aging. Though such obstacles are addressed and partially overcome for embryonic premature and mature CNS tissues, procedural adaptations to an aged, progeroid, and degenerative CNS environment are underrepresented. Here, we describe a practical workflow for the acquisition and phenomapping of CNS neural cells at states of health, physiological and precocious aging, and genetically provoked neurodegeneration. Following recent, unprecedented evidence of post-mitotic cellular senescence (PoMiCS), the protocol appears suitable for such de novo characterization and phenotypic opposition to classical senescence. Technically, the protocol is rapid, efficient as for cellular yield and well preserves physiological cell proportions. It is suitable for a variety of downstream applications aiming at cell type-specific interrogations, including cell culture systems, Flow-FISH, flow cytometry/FACS, senescence studies, and retrieval of omic-scale DNA, RNA, and protein profiles. We expect suitability for transfer to other CNS targets and to a broad spectrum of engineered systems addressing aging, neurodegeneration, progeria, and senescence.
Collapse
Affiliation(s)
- Quratul Ain
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany; (C.W.S.); (D.W.); (O.W.W.)
- Correspondence: (Q.A.); (A.K.); Tel.: +49-3641-9396630 (Q.A.); +49-3641-9323499 (A.K.)
| | - Christian W. Schmeer
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany; (C.W.S.); (D.W.); (O.W.W.)
| | - Diane Wengerodt
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany; (C.W.S.); (D.W.); (O.W.W.)
| | - Yvonne Hofmann
- Department of Internal Medicine V, Jena University Hospital, 07747 Jena, Germany;
| | - Otto W. Witte
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany; (C.W.S.); (D.W.); (O.W.W.)
| | - Alexandra Kretz
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany; (C.W.S.); (D.W.); (O.W.W.)
- Correspondence: (Q.A.); (A.K.); Tel.: +49-3641-9396630 (Q.A.); +49-3641-9323499 (A.K.)
| |
Collapse
|
49
|
Neurogenetic disorders across the lifespan: from aberrant development to degeneration. Nat Rev Neurol 2022; 18:117-124. [PMID: 34987232 PMCID: PMC10132523 DOI: 10.1038/s41582-021-00595-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 02/08/2023]
Abstract
Intellectual disability and autism spectrum disorder (ASD) are common, and genetic testing is increasingly performed in individuals with these diagnoses to inform prognosis, refine management and provide information about recurrence risk in the family. For neurogenetic conditions associated with intellectual disability and ASD, data on natural history in adults are scarce; however, as older adults with these disorders are identified, it is becoming clear that some conditions are associated with both neurodevelopmental problems and neurodegeneration. Moreover, emerging evidence indicates that some neurogenetic conditions associated primarily with neurodegeneration also affect neurodevelopment. In this Perspective, we discuss examples of diseases that have developmental and degenerative overlap. We propose that neurogenetic disorders should be studied continually across the lifespan to understand the roles of the affected genes in brain development and maintenance, and to inform strategies for treatment.
Collapse
|
50
|
Hickman RA, Dewan R, Cortes E, Traynor BJ, Marder K, Vonsattel JP. Amyotrophic lateral sclerosis is over-represented in two Huntington's disease brain bank cohorts: further evidence to support genetic pleiotropy of pathogenic HTT gene expansion. Acta Neuropathol 2022; 143:105-108. [PMID: 34800149 PMCID: PMC8918027 DOI: 10.1007/s00401-021-02385-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 12/28/2022]
Affiliation(s)
- Richard A Hickman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 630 W 168th Street, New York, NY, 10032, USA.
| | - Ramita Dewan
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, 20892, USA
| | - Etty Cortes
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, 20892, USA
| | - Karen Marder
- Department of Neurology and Psychiatry, Columbia University Irving Medical Center, New York, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 710 West 168th Street, New York, NY, 10032, USA
| | - Jean-Paul Vonsattel
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 630 W 168th Street, New York, NY, 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 710 West 168th Street, New York, NY, 10032, USA
| |
Collapse
|