1
|
van den Bos MAJ, Menon P, Pavey N, Higashihara M, Kiernan MC, Vucic S. Direct interrogation of cortical interneuron circuits in amyotrophic lateral sclerosis. Brain 2025; 148:1169-1179. [PMID: 39385724 DOI: 10.1093/brain/awae317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/30/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Cortical hyperexcitability is a key pathogenic feature of amyotrophic lateral sclerosis (ALS), believed to be mediated through complex interplay of cortical interneurons. To date, there has been no technological approach to facilitate the direct capture of cortical interneuron function. Through combination of transcranial magnetic stimulation (TMS) with advanced EEG, the present study examined GABAergic dysfunction in ALS by recording focused cortical output whilst applying TMS over the primary motor cortex contralateral to the site of symptom onset. Using both a single-pulse and a novel inhibitory paired-pulse paradigm, TMS-EEG studies were undertaken on 21 ALS patients and results compared with healthy controls. TMS responses captured by EEG form a discrete waveform known as the transcranial evoked potential (TEP), with positive (P) or upward deflections occurring at 30 (P30), 60 (P60) and 190 ms (P190) after TMS stimulus. Negative (N) or downward deflections occur at 44 (N44), 100 (N100) and 280 ms (N280) after TMS stimulus. The single-pulse TEPs recorded in ALS patients demonstrated novel differences suggestive of cortical GABAergic dysfunction. When compared with controls, the N100 component was significantly reduced (P < 0.05), whereas the P190 component increased (P < 0.05) in ALS patients. Additionally, the N44 component was correlated with muscle weakness (r = -0.501, P < 0.05). These findings were supported by reduced paired-pulse inhibition of TEP components in ALS patients (P60, P < 0.01; N100, P < 0.005), consistent with dysfunction of cortical interneuronal GABAA-ergic circuits. Furthermore, the reduction in short-interval intracortical inhibition, as reflected by changes in paired-pulse inhibition of the N100 component, was associated with longer disease duration in ALS patients (r = -0.698, P < 0.001). In conclusion, intensive and focused interrogation of the motor cortex using novel TMS-EEG combined technologies has established localized dysfunction of GABAergic circuits, supporting the notion that cortical hyperexcitability is mediated by cortical disinhibition in ALS. Dysfunction of GABAergic circuits was correlated with greater clinical disability and disease duration, implying pathophysiological significance.
Collapse
Affiliation(s)
- Mehdi A J van den Bos
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Parvathi Menon
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Nathan Pavey
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Matthew C Kiernan
- Neuroscience Research Australia, Randwick, Sydney, NSW 2031, Australia
- Institute of Neurological Sciences, Eastern Sydney Local Health District, Randwick, Sydney, NSW 2031, Australia
| | - Steve Vucic
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Sydney, NSW 2139, Australia
- Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| |
Collapse
|
2
|
D'Amico A, Cucunato R, Salemi G, Bella VL, Aridon P. A population based study to analyse amyotrophic lateral sclerosis as a multi-step process. Sci Rep 2025; 15:11189. [PMID: 40169635 PMCID: PMC11962139 DOI: 10.1038/s41598-025-89616-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Recent studies suggest that Amyotrophic Lateral Sclerosis (ALS) follows a multistep process. We evaluated this hypothesis in a well-defined ALS population in Palermo, Sicily, almost entirely followed by our ALS Clinical Center. Incident data from the ALS Center (2014-2023) were analyzed, including both sporadic and familial ALS forms of the disease. To evaluate the multistep process, we regressed the natural log of age-specific incidence against the natural log of patient age We identified 216 ALS patients. We obtained a slope of 5 (r2 = 0.93); the 95% CI ranged from 2.51 to 7.60, remaining relatively wide due to the small sample size, with a p-value of 0.008. The slope estimate was consistent with a 6-step process. In the Palermo ALS population, the multistep analysis confirms a process consistent with a 6-step model. This data, obtained in a relatively homogeneous population, further highlights the probability of strict interaction between environmental and genetic variables in the disease. Our data offer insights into the complexity of the mechanisms involved in the pathogenesis of the disease, particularly during its asymptomatic phase. This study supports the hypothesis that a single therapeutic silver bullet would probably be insufficient to arrest or slow the disease's progression.
Collapse
Affiliation(s)
- Anna D'Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, Palermo, 90129, Italy
| | - Roberta Cucunato
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, Palermo, 90129, Italy
| | - Giuseppe Salemi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, Palermo, 90129, Italy
| | - Vincenzo La Bella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, Palermo, 90129, Italy
| | - Paolo Aridon
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, Palermo, 90129, Italy.
| |
Collapse
|
3
|
Carroll E, Scaber J, Huber KVM, Brennan PE, Thompson AG, Turner MR, Talbot K. Drug repurposing in amyotrophic lateral sclerosis (ALS). Expert Opin Drug Discov 2025; 20:447-464. [PMID: 40029669 PMCID: PMC11974926 DOI: 10.1080/17460441.2025.2474661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Identifying treatments that can alter the natural history of amyotrophic lateral sclerosis (ALS) is challenging. For years, drug discovery in ALS has relied upon traditional approaches with limited success. Drug repurposing, where clinically approved drugs are reevaluated for other indications, offers an alternative strategy that overcomes some of the challenges associated with de novo drug discovery. AREAS COVERED In this review, the authors discuss the challenge of drug discovery in ALS and examine the potential of drug repurposing for the identification of new effective treatments. The authors consider a range of approaches, from screening in experimental models to computational approaches, and outline some general principles for preclinical and clinical research to help bridge the translational gap. Literature was reviewed from original publications, press releases and clinical trials. EXPERT OPINION Despite remaining challenges, drug repurposing offers the opportunity to improve therapeutic options for ALS patients. Nevertheless, stringent preclinical research will be necessary to identify the most promising compounds together with innovative experimental medicine studies to bridge the translational gap. The authors further highlight the importance of combining expertise across academia, industry and wider stakeholders, which will be key in the successful delivery of repurposed therapies to the clinic.
Collapse
Affiliation(s)
- Emily Carroll
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jakub Scaber
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Kilian V. M. Huber
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Paul E. Brennan
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Ye L, Dittlau KS, Sicart A, Janky R, Van Damme P, Van Den Bosch L. Sporadic ALS hiPSC-derived motor neurons show axonal defects linked to altered axon guidance pathways. Neurobiol Dis 2025; 206:106815. [PMID: 39884586 DOI: 10.1016/j.nbd.2025.106815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder characterized by the selective and progressive loss of motor neurons, leading to gradual paralysis and death within 2 to 5 years after diagnosis. The exact underlying pathogenic mechanism(s) remain elusive. This is particularly the case for sporadic ALS (sALS), representing 90 % of cases, as modelling a sporadic disease is extremely difficult. We used human induced pluripotent stem cell (hiPSC)-derived motor neurons from sALS patients to investigate early disease mechanisms. The earliest phenotype that we observed were profound axonal defects including impaired axonal transport, defective axonal outgrowth and a reduced formation of neuromuscular junctions. Transcriptomic profiling revealed significant dysregulation in axon guidance pathways, with upregulation of specific axonal regeneration-inhibiting genes, such as EphA4 and DCC in sALS motor neurons. Our findings suggest that dysregulation of axon guidance pathways contributes to axonal defects and that this could play a crucial role in the pathogenesis of sALS.
Collapse
Affiliation(s)
- Lisha Ye
- KU Leuven - University of Leuven, Department of Neurosciences and Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Katarina Stoklund Dittlau
- KU Leuven - University of Leuven, Department of Neurosciences and Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Adria Sicart
- KU Leuven - University of Leuven, Department of Neurosciences and Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium
| | | | - Philip Van Damme
- KU Leuven - University of Leuven, Department of Neurosciences and Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium; University Hospitals Leuven, Department of Neurology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences and Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium.
| |
Collapse
|
5
|
Dellar ER, Vendrell I, Amein B, Lester DG, Edmond EC, Yoganathan K, Dharmadasa T, Sogorb‐Esteve A, Fischer R, Talbot K, Rohrer JD, Turner MR, Thompson AG. Elevated Cerebrospinal Fluid Ubiquitin Carboxyl-Terminal Hydrolase Isozyme L1 in Asymptomatic C9orf72 Hexanucleotide Repeat Expansion Carriers. Ann Neurol 2025; 97:449-459. [PMID: 39548852 PMCID: PMC11831881 DOI: 10.1002/ana.27133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/29/2024] [Accepted: 10/25/2024] [Indexed: 11/18/2024]
Abstract
OBJECTIVE To identify biochemical changes in individuals at higher risk of developing amyotrophic lateral sclerosis (ALS) or frontotemporal dementia (FTD) via C9orf72 hexanucleotide repeat expansion (HRE) heterozygosity. METHODS Cross-sectional observational study of 48 asymptomatic C9orf72 HRE carriers, 39 asymptomatic non-carrier controls, 19 people with sporadic ALS, 10 with C9orf72 ALS, 14 with sporadic FTD, and 10 with C9orf72 FTD. Relative abundance of 30 pre-defined cerebrospinal fluid biomarkers of ALS and FTD were compared in asymptomatic C9orf72 HRE carriers and age-matched non-carrier controls. Differential abundance of these proteins was quantified using data independent acquisition mass spectrometry or electro chemiluminescent assay for neurofilament light chain. Unbiased analysis of the entire cerebrospinal fluid proteome was then carried out. RESULTS Ubiquitin carboxyl-hydrolase isozyme L1 levels were higher in asymptomatic C9orf72 HRE carriers compared with age-matched non-carriers (log2fold change 0.20, FDR-adjusted p-value = 0.034), whereas neurofilament light chain levels did not significantly differ. Ubiquitin carboxyl-hydrolase isozyme L1 levels remained elevated after matching of groups by neurofilament levels (p = 0.011), and after adjusting for age, sex, and neurofilament levels. A significant difference was also observed when restricting analysis to younger participants (<37) matched by neurofilament level (p = 0.007). INTERPRETATION Elevated cerebrospinal fluid ubiquitin carboxyl-hydrolase isozyme L1 levels in C9orf72 HRE carriers can occur in the absence of increased neurofilament levels, potentially reflecting either compensatory or pathogenic mechanisms preceding rapid neuronal loss. This brings forward the window on changes associated with the C9orf72 HRE carrier state, with potential to inform understanding of penetrance and approaches to prevention. ANN NEUROL 2025;97:449-459.
Collapse
Affiliation(s)
| | - Iolanda Vendrell
- Target Discovery InstituteCentre for Medicines Discovery, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUK
| | - Benazir Amein
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - David G. Lester
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Evan C. Edmond
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Katie Yoganathan
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Thanuja Dharmadasa
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkville, VictoriaAustralia
| | - Aitana Sogorb‐Esteve
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research Institute at University College LondonLondonUK
| | - Roman Fischer
- Target Discovery InstituteCentre for Medicines Discovery, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUK
| | - Kevin Talbot
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Jonathan D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Martin R. Turner
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | | |
Collapse
|
6
|
Eisen A, Kiernan MC. The Neonatal Microbiome: Implications for Amyotrophic Lateral Sclerosis and Other Neurodegenerations. Brain Sci 2025; 15:195. [PMID: 40002527 PMCID: PMC11852589 DOI: 10.3390/brainsci15020195] [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: 01/13/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Most brain development occurs in the "first 1000 days", a critical period from conception to a child's second birthday. Critical brain processes that occur during this time include synaptogenesis, myelination, neural pruning, and the formation of functioning neuronal circuits. Perturbations during the first 1000 days likely contribute to later-life neurodegenerative disease, including sporadic amyotrophic lateral sclerosis (ALS). Neurodevelopment is determined by many events, including the maturation and colonization of the infant microbiome and its metabolites, specifically neurotransmitters, immune modulators, vitamins, and short-chain fatty acids. Successful microbiome maturation and gut-brain axis function depend on maternal factors (stress and exposure to toxins during pregnancy), mode of delivery, quality of the postnatal environment, diet after weaning from breast milk, and nutritional deficiencies. While the neonatal microbiome is highly plastic, it remains prone to dysbiosis which, once established, may persist into adulthood, thereby inducing the development of chronic inflammation and abnormal excitatory/inhibitory balance, resulting in neural excitation. Both are recognized as key pathophysiological processes in the development of ALS.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Matthew C. Kiernan
- Neuroscience Research Australia, University of New South Wales, Randwick, Sydney, NSW 2031, Australia;
| |
Collapse
|
7
|
van Veenhuijzen K, Tan HH, Nitert AD, van Es MA, Veldink JH, van den Berg LH, Westeneng H. Longitudinal Magnetic Resonance Imaging in Asymptomatic C9orf72 Mutation Carriers Distinguishes Phenoconverters to Amyotrophic Lateral Sclerosis or Amyotrophic Lateral Sclerosis With Frontotemporal Dementia. Ann Neurol 2025; 97:281-295. [PMID: 39487710 PMCID: PMC11740280 DOI: 10.1002/ana.27116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 10/01/2024] [Accepted: 10/05/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVE We prospectively studied asymptomatic C9orf72 mutation carriers, identifying those developing amyotrophic lateral sclerosis (ALS) or frontotemporal dementia (FTD). METHODS We enrolled 56 asymptomatic family members (AFM) with a C9orf72 mutation (AFM C9+), 132 non-carriers (AFM C9-), and 359 population-based controls. Using 3 T magnetic resonance imaging, we measured cortical thickness, gyrification, and subcortical volumes longitudinally. Linear mixed-effects models on non-converting AFM C9+ scans (n = 107) created a reference for these measurements, establishing individual atrophy patterns. Atrophy patterns from presymptomatic phenoconverters (n = 10 scans) served as a template for group comparisons and similarity assessments. Similarity with phenoconverters was quantified using Dice similarity coefficient (DSC) for cortical and Kullback-Leibler similarity (KLS) for subcortical measures. Using longitudinal similarity assessments, we predicted when participants would reach the average similarity level of phenoconverters at their first post-onset scan. RESULTS Five AFM C9+ converted to ALS or ALS-FTD. Up to 6 years before symptoms, these phenoconverters exhibited significant atrophy in frontal, temporal, parietal, and cingulate cortex, along with smaller thalamus, hippocampus, and amygdala compared to other AFM C9+. Some non-converted AFM C9+ had high DSC and KLS, approaching values of phenoconverters, whereas others, along with AFM C9- and controls, had lower values. At age 80, we predicted 27.9% (95% confidence interval, 13.2-40.1%) of AFM C9+ and no AFM C9- would reach the same DSC as phenoconverters. INTERPRETATION Distinctive atrophy patterns are visible years before symptom onset on presymptomatic scans of phenoconverters. Combining baseline and follow-up similarity measures may serve as a promising imaging biomarker for identifying those at risk of ALS or ALS-FTD. ANN NEUROL 2025;97:281-295.
Collapse
Affiliation(s)
- Kevin van Veenhuijzen
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Harold H.G. Tan
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Abram D. Nitert
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Michael A. van Es
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Jan H. Veldink
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Leonard H. van den Berg
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Henk‐Jan Westeneng
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
8
|
Devarajan A, Seah C, Zhang JY, Vasan V, Feng R, Chapman EK, Shigematsu T, Bederson J, Shrivastava RK. A four-hit mechanism is sufficient for meningioma development. J Neurooncol 2025; 171:599-607. [PMID: 39586894 DOI: 10.1007/s11060-024-04877-y] [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: 10/12/2024] [Accepted: 11/01/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE Meningiomas are central nervous system tumors whose incidence increases with age. Benign meningioma pathogenesis involves germline or somatic mutation of target genes, such as NF2, leading to clonal expansion. We used an established cancer epidemiology model to investigate the number of rate-limiting steps sufficient for benign meningioma development. METHODS Incidence data was obtained from the Surveillance, Epidemiology and End Results Program (SEER) for nonmalignant meningioma from 2004 to 2020. Age-adjusted incidence rates per 100,000 person-years were divided into 5-year bands. This was repeated for vestibular schwannomas as a negative control. The Armitage-Doll methodology was applied. Mathematical solutions correcting for volatile tumor microenvironments were applied to fit higher-order models using polynomial regression when appropriate. A 75:25 training:test split was utilized for validation. RESULTS 222,509 cases of benign meningiomas were identified. We noted strong linear relationships between log-transformed incidence and age across the cohort and multiple subpopulations: male, white, black, Hispanic, Asian/Pacific Islander, and American Indian subpopulations all demonstrated R2 = 0.99. Slopes were between 3.1 and 3.4, suggesting a four-step process for benign meningioma development. Female patients exhibited nonlinear deviations, but the corrected model demonstrated R2 = 0.99 with a four-hit pathway. This model performed robustly on test data with R2 = 0.99. Vestibular schwannomas demonstrated a slope of 2.1 with R2 = 0.99, suggesting a separate three-step process. CONCLUSION Four mutations are uniquely required for the development of benign meningiomas. Correcting for volatile tumor microenvironments reliably accounted for nonlinear deviations in behavior. Further studies are warranted to elucidate genomic findings suggestive of key mutations in this pathway.
Collapse
Affiliation(s)
- Alex Devarajan
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Carina Seah
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack Y Zhang
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vikram Vasan
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rui Feng
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily K Chapman
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tomoyoshi Shigematsu
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua Bederson
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raj K Shrivastava
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
9
|
Awuah WA, Shah MH, Sanker V, Mannan KM, Ranganathan S, Nkrumah-Boateng PA, Frimpong M, Darko K, Tan JK, Abdul-Rahman T, Atallah O. Advances in chromosomal microarray analysis: Transforming neurology and neurosurgery. BRAIN & SPINE 2025; 5:104197. [PMID: 39990116 PMCID: PMC11847126 DOI: 10.1016/j.bas.2025.104197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/25/2025]
Abstract
Over the past two decades, genomics has transformed our understanding of various clinical conditions, with Chromosomal Microarray Analysis (CMA) standing out as a key technique. Offering unparalleled sensitivity, CMA detects submicroscopic chromosomal imbalances, enabling the examination of DNA for copy number variations, deletions, duplications, and other structural differences. In neurology, CMA has revolutionised diagnoses, personalised treatment plans, and patient outcomes. By identifying genetic anomalies linked to neurological conditions, CMA allows clinicians to tailor treatments based on individual genetic profiles, enhancing precision medicine. CMA's clinical utility spans numerous neurological conditions, providing crucial insights into neurodevelopmental disorders, CNS tumours, neurodegenerative diseases, cerebrovascular diseases, and epilepsy. In neurodevelopmental disorders, CMA aids in diagnosing autism and intellectual disabilities, facilitating early interventions that improve long-term outcomes. In epilepsy, CMA helps identify genetic causes of drug-resistant seizures, enabling more targeted therapies and reducing adverse reactions. CMA also aids in stratifying risk for cerebrovascular diseases, enabling preventive interventions that improve patient prognosis. Despite its potential, challenges remain, such as interpreting variants of uncertain significance (VOUS), the lack of standardised testing guidelines, and issues of cost and accessibility. Addressing these challenges will optimise CMA's impact, advancing personalised medicine and reshaping neurology. This review discusses CMA's pivotal role in bridging the gap between genomics and clinical practice, underscoring its potential to transform neurogenetics and ultimately improve patient care.
Collapse
Affiliation(s)
| | | | - Vivek Sanker
- Department of Neurosurgery, Trivandrum Medical College, India
- Department of Neurosurgery, Stanford University, CA, USA
| | | | - Sruthi Ranganathan
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Mabel Frimpong
- Bryn Mawr College, 101 N Merion Avenue, Bryn Mawr, PA, USA
| | - Kwadwo Darko
- Department of Neurosurgery, Korle Bu Teaching Hospital, Accra, Ghana
| | - Joecelyn Kirani Tan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | | | - Oday Atallah
- Department of Neurosurgery, Carl Von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
10
|
Trojsi F, Canna A, Sharbafshaaer M, di Nardo F, Canale F, Passaniti C, Pirozzi MA, Silvestro M, Orologio I, Russo A, Cirillo M, Tessitore A, Siciliano M, Esposito F. Brain neurovascular coupling in amyotrophic lateral sclerosis: Correlations with disease progression and cognitive impairment. Eur J Neurol 2025; 32:e16540. [PMID: 39529471 PMCID: PMC11625914 DOI: 10.1111/ene.16540] [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: 08/18/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND PURPOSE 'Neurovascular coupling' (NVC) alterations, assessing the interplay between local cerebral perfusion and neural activity within a given brain region or network, may reflect neurovascular unit impairment in amyotrophic lateral sclerosis (ALS). The aim was to explore NVC as a correlation between the functional connectivity and cerebral blood flow within the large-scale resting-state functional magnetic resonance imaging brain networks in a sample of ALS patients compared to healthy controls (HCs). METHODS Forty-eight ALS patients (30 males; mean age 60.64 ± 9.62 years) and 32 HC subjects (14 males; mean age 55.06 ± 16 years) were enrolled and underwent 3 T magnetic resonance imaging. ALS patients were screened by clinical and neuropsychological scales and were retrospectively classified as very fast progressors (VFPs), fast progressors and slow progressors (SPs). RESULTS Neurovascular coupling reduction within the default mode network (DMN) (p = 0.005) was revealed in ALS patients compared to HCs, observing, for this network, significant NVC differences between VFP and SP groups. Receiver operating characteristic curve analysis showed that impaired NVC in the DMN at baseline best discriminated VFPs and SPs (area under the curve 75%). Significant correlations were found between NVC and the executive (r = 0.40, p = 0.01), memory (r = 0.32, p = 0.04), visuospatial ability (r = 0.40, p = 0.01) and non-ALS-specific (r = 0.40, p = 0.01) subscores of the Edinburgh Cognitive and Behavioural ALS Screen. CONCLUSIONS The reduction of brain NVC in the DMN may reflect largely distributed abnormalities of the neurovascular unit. NVC alterations in the DMN could play a role in anticipating a faster clinical progression in ALS patients, aiding patient selection and monitoring during clinical trials.
Collapse
Affiliation(s)
- Francesca Trojsi
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
- First Division of Neurology and NeurophysiopathologyAOU Università degli Studi della Campania ‘Luigi Vanvitelli’NaplesItaly
| | - Antonietta Canna
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
- Montreal Neurological Institute and Hospital, McGill UniversityMontrealQuebecCanada
| | - Minoo Sharbafshaaer
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
| | - Federica di Nardo
- First Division of Neurology and NeurophysiopathologyAOU Università degli Studi della Campania ‘Luigi Vanvitelli’NaplesItaly
| | - Fabrizio Canale
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
- First Division of Neurology and NeurophysiopathologyAOU Università degli Studi della Campania ‘Luigi Vanvitelli’NaplesItaly
| | - Carla Passaniti
- First Division of Neurology and NeurophysiopathologyAOU Università degli Studi della Campania ‘Luigi Vanvitelli’NaplesItaly
| | - Maria Agnese Pirozzi
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
| | - Marcello Silvestro
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
| | - Ilaria Orologio
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
- First Division of Neurology and NeurophysiopathologyAOU Università degli Studi della Campania ‘Luigi Vanvitelli’NaplesItaly
| | - Antonio Russo
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
- First Division of Neurology and NeurophysiopathologyAOU Università degli Studi della Campania ‘Luigi Vanvitelli’NaplesItaly
| | - Mario Cirillo
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
| | - Alessandro Tessitore
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
- First Division of Neurology and NeurophysiopathologyAOU Università degli Studi della Campania ‘Luigi Vanvitelli’NaplesItaly
| | - Mattia Siciliano
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
- Department of PsychologyUniversità degli Studi della Campania ‘Luigi Vanvitelli’CasertaItaly
- Neurosciences Research CentreMolecular and Clinical Sciences Research Institute, St George's, University of LondonLondonUK
| | - Fabrizio Esposito
- Department of Advanced Medical and Surgical SciencesMRI Research Center, Università degli Studi della Campania Luigi VanvitelliNaplesItaly
| |
Collapse
|
11
|
Ziser L, van Eijk RPA, Kiernan MC, McRae A, Henderson RD, Schultz D, Needham M, Mathers S, McCombe P, Talman P, Vucic S. Amyotrophic lateral sclerosis established as a multistep process across phenotypes. Eur J Neurol 2025; 32:e16532. [PMID: 39475283 PMCID: PMC11622508 DOI: 10.1111/ene.16532] [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: 08/06/2024] [Revised: 09/21/2024] [Accepted: 10/07/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND PURPOSE Given the accepted multistep process of disease causation in amyotrophic lateral sclerosis (ALS), the present study was undertaken to determine the number of steps required for disease onset across each of the ALS phenotypes. METHODS Clinical and demographic data were prospectively accumulated using the Australian Motor Neurone Disease Registry (2005-2016), and age-specific incidence rates were calculated. Poisson regression was utilized to assess the relationship between log age-specific incidence and log age of onset, with McFadden's R2 used to assess the goodness of fit of the model. RESULTS In total, 2647 ALS patients were included, with mean disease-onset age being 62.2 ± 12.1 years. A linear relationship between log incidence and log age was established across ALS phenotypes, with variable slope estimates: bulbar 5.1 (95% confidence interval [CI] 4.6-5.6); cervical 2.7 (95% CI 2.3-3.0); lumbar 3.5 (95% CI 3.2-3.9); flail arm 4.7 (95% CI 3.9-5.5); flail leg 3.6 (95% CI 2.6-4.5); primary lateral sclerosis 2.7 (95% CI 1.8-3.7). Slope estimates were significantly higher in the bulbar compared to the cervical, lumbar and primary lateral sclerosis phenotypes. McFadden's R2 values were >0.4 for all phenotypes indicating excellent model fit. DISCUSSION A multistep process has been established across all ALS phenotypes with variable slope estimates, suggesting that the number of steps to develop disease is different across clinical presentations. Identification of mechanisms underlying slope estimate variability could exert pathophysiological significance.
Collapse
Affiliation(s)
- Laura Ziser
- Institute for Molecular Bioscience, University of QueenslandBrisbaneQueenslandAustralia
| | - Ruben P. A. van Eijk
- Department of Neurology, UMC Utrecht Brain CentreUniversity Medical Center UtrechtUtrechtThe Netherlands
- Biostatistics and Research Support, Julius Centre for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Allan McRae
- Institute for Molecular Bioscience, University of QueenslandBrisbaneQueenslandAustralia
| | - Robert D. Henderson
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - David Schultz
- Department of NeurologyFlinders University of South Australia, and Flinders Medical CentreBedford ParkSouth AustraliaAustralia
| | - Merrilee Needham
- Department of NeurologyFiona Stanley HospitalMurdochWestern AustraliaAustralia
- Centre for Molecular, MedicineMurdoch UniversityPerthWestern AustraliaAustralia
- Department of NeurologyPerron Institute for Neurological and Translational ScienceNedlandsWestern AustraliaAustralia
- Department of NeurologyUniversity of Notre DameFremantleWestern AustraliaAustralia
| | - Susan Mathers
- Department of NeurologyCalvary Health Care BethlehemMelbourneVictoriaAustralia
- School of Clinical Sciences, Monash UniversityMelbourneVictoriaAustralia
| | - Pam McCombe
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Paul Talman
- Deakin University, University Hospital GeelongGeelongVictoriaAustralia
| | - Steve Vucic
- Brain and Nerve Research CentreThe University of Sydney, Concord HospitalSydneyNew South WalesAustralia
| |
Collapse
|
12
|
Cocoș R, Popescu BO. Scrutinizing neurodegenerative diseases: decoding the complex genetic architectures through a multi-omics lens. Hum Genomics 2024; 18:141. [PMID: 39736681 DOI: 10.1186/s40246-024-00704-7] [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: 10/05/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
Neurodegenerative diseases present complex genetic architectures, reflecting a continuum from monogenic to oligogenic and polygenic models. Recent advances in multi-omics data, coupled with systems genetics, have significantly refined our understanding of how these data impact neurodegenerative disease mechanisms. To contextualize these genetic discoveries, we provide a comprehensive critical overview of genetic architecture concepts, from Mendelian inheritance to the latest insights from oligogenic and omnigenic models. We explore the roles of common and rare genetic variants, gene-gene and gene-environment interactions, and epigenetic influences in shaping disease phenotypes. Additionally, we emphasize the importance of multi-omics layers including genomic, transcriptomic, proteomic, epigenetic, and metabolomic data in elucidating the molecular mechanisms underlying neurodegeneration. Special attention is given to missing heritability and the contribution of rare variants, particularly in the context of pleiotropy and network pleiotropy. We examine the application of single-cell omics technologies, transcriptome-wide association studies, and epigenome-wide association studies as key approaches for dissecting disease mechanisms at tissue- and cell-type levels. Our review introduces the OmicPeak Disease Trajectory Model, a conceptual framework for understanding the genetic architecture of neurodegenerative disease progression, which integrates multi-omics data across biological layers and time points. This review highlights the critical importance of adopting a systems genetics approach to unravel the complex genetic architecture of neurodegenerative diseases. Finally, this emerging holistic understanding of multi-omics data and the exploration of the intricate genetic landscape aim to provide a foundation for establishing more refined genetic architectures of these diseases, enhancing diagnostic precision, predicting disease progression, elucidating pathogenic mechanisms, and refining therapeutic strategies for neurodegenerative conditions.
Collapse
Affiliation(s)
- Relu Cocoș
- Department of Medical Genetics, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.
- Genomics Research and Development Institute, Bucharest, Romania.
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.
| |
Collapse
|
13
|
Pamphlett R, Parkin Kullmann J. Early life events may be the first steps on the multistep path to amyotrophic lateral sclerosis. Sci Rep 2024; 14:28497. [PMID: 39557859 PMCID: PMC11573994 DOI: 10.1038/s41598-024-78240-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/29/2024] [Indexed: 11/20/2024] Open
Abstract
A combination of multiple genetic and environmental factors appear to be required to trigger the onset of amyotrophic lateral sclerosis (ALS). Early life environmental exposures have been reported to be risk factors for a variety of adult-onset diseases, so we used data from an online international ALS case-control questionnaire to estimate whether any of these could be risk factors for the clinical onset of ALS. Responses were obtained from 1,049 people aged 40 years or more, 568 with ALS and 481 controls. People with ALS were more likely to have been born and lived longer in a country area than in a city area, to have younger parents, and to have lower educational attainment and fewer years of education. No ALS-control differences were found in sibling numbers, birth order, adult height, birth weight, parent smoking, Cesarean delivery, or age of starting smoking. In conclusion, early life events and conditions may be part of a group of polyenvironmental risk factors that act together with polygenetic variants to trigger the onset of ALS. Reducing exposure to adverse environmental factors in early life could help to lower the risk of later developing ALS.
Collapse
Affiliation(s)
- Roger Pamphlett
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
- Department of Pathology, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia.
| | - Jane Parkin Kullmann
- Department of Pathology, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
14
|
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
|
15
|
Theme 2 Genetics and Genomics. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:105-121. [PMID: 39508667 DOI: 10.1080/21678421.2024.2403299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
|
16
|
Theme 1 Epidemiology and Informatics. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:93-104. [PMID: 39508668 DOI: 10.1080/21678421.2024.2403298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
|
17
|
Mirceta M, Schmidt MHM, Shum N, Prasolava TK, Meikle B, Lanni S, Mohiuddin M, Mckeever PM, Zhang M, Liang M, van der Werf I, Scheers S, Dion PA, Wang P, Wilson MD, Abell T, Philips EA, Sznajder ŁJ, Swanson MS, Mehkary M, Khan M, Yokoi K, Jung C, de Jong PJ, Freudenreich CH, McGoldrick P, Yuen RKC, Abrahão A, Keith J, Zinman L, Robertson J, Rogaeva E, Rouleau GA, Kooy RF, Pearson CE. C9orf72 expansion creates the unstable folate-sensitive fragile site FRA9A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.26.620312. [PMID: 39569145 PMCID: PMC11577248 DOI: 10.1101/2024.10.26.620312] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
The hyper-unstable Chr9p21 locus, harbouring the interferon gene cluster, oncogenes and C9orf72, is linked to multiple diseases. C9orf72 (GGGGCC)n expansions ( C9orf72 Exp) are associated with incompletely penetrant amyotrophic lateral sclerosis, frontotemporal dementia and autoimmune disorders. C9orf72 Exp patients display hyperactive cGAS-STING-linked interferon immune and DNA damage responses, but the source of immuno-stimulatory or damaged DNA is unknown. Here, we show C9orf72 Exp in pre-symptomatic and ALS-FTD patient cells and brains cause the folate-sensitive chromosomal fragile site, FRA9A. FRA9A centers on >33kb of C9orf72 as highly-compacted chromatin embedded in an 8.2Mb fragility zone spanning 9p21, encompassing 46 genes, making FRA9A one of the largest fragile sites. C9orf72 Exp cells show chromosomal instability, heightened global- and Chr9p-enriched sister-chromatid exchanges, truncated-Chr9s, acentric-Chr9s and Chr9-containing micronuclei, providing endogenous sources of damaged and immunostimulatory DNA. Cells from one C9orf72 Exp patient contained highly-rearranged FRA9A-expressing Chr9 with Chr9-wide dysregulated gene expression. Somatic C9orf72 Exp repeat instability and chromosomal fragility are sensitive to folate-deficiency. Age-dependent repeat instability, chromosomal fragility, and chromosomal instability can be transferred to CNS and peripheral tissues of transgenic C9orf72 Exp mice, implicating C9orf72 Exp as the source. Our results highlight unappreciated effects of C9orf72 expansions that trigger vitamin-sensitive chromosome fragility, adding structural variations to the disease-enriched 9p21 locus, and likely elsewhere.
Collapse
|
18
|
Santos Silva C, Gormicho M, Simão S, Pronto-Laborinho AC, Alves I, Pinto S, Oliveira Santos M, de Carvalho M. C9orf72 gene repeat expansion phenotype profile of motor neurone disease in Portugal. J Neurol Sci 2024; 465:123208. [PMID: 39226712 DOI: 10.1016/j.jns.2024.123208] [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: 05/20/2024] [Revised: 07/18/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND C9orf72 gene repeat expansion (C9RE) is the most frequent gene variant associated with amyotrophic lateral sclerosis (ALS). We aimed to study the phenotype of motor neurone disease (MND) patients with C9RE in a Portuguese cohort. METHODS Demographical and clinical data of MND patients with (C9RE+) and without C9RE were compared. ALS al Rating Scale-Revised (ALSFRS-R) and Edinburgh Cognitive and Behavioural ALS Screen (ECAS) were used to evaluate functional and cognitive performance, respectively. Survival analysis was performed using Kaplan Meier log-rank test and Cox proportional hazards model. RESULTS We included 761 patients of whom 61 (8.0 %) were C9RE+. C9RE+ patients had a higher frequency of ALS (95.1 vs 78.4 %, p = 0.002), and lower frequency of progressive muscular atrophy (3.3 vs 16.7 %, p = 0.006). C9RE+ was associated with earlier age of onset (58.1 vs 62.6 years, p = 0.003) and more frequent MND family history (65.5 vs 11.4 %, p < 0.001). Gender, ethnicity, onset site, diagnostic delay, disease progression rate until diagnosis (ΔF), ALSFRS-R and time until non-invasive ventilation did not differ between groups. Cognitive/behavioural symptoms and ECAS did not differ between groups, except a worse visuospatial score in C9RE+ group (p = 0.035). Death rate was 1.8 and 1.6 times higher in C9RE+ patients with MND and ALS, respectively. Significant survival prognostic factors in C9RE+ group were diagnosis delay (HR = 0.96, 95 %CI 0.92-0.99, p = 0.008) and ΔF (HR = 1.93, 95 %CI 1.26-2.96, p = 0.002). CONCLUSION Our study corroborates most previous cohorts' findings, but harbours some singularities regarding onset site, phenotype, and cognitive profile, that contribute to a better understanding of C9RE epidemiology.
Collapse
Affiliation(s)
- Cláudia Santos Silva
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal; Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal.
| | - Marta Gormicho
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Simão
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Catarina Pronto-Laborinho
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Inês Alves
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Susana Pinto
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Oliveira Santos
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal; Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| | - Mamede de Carvalho
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal; Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
19
|
Cheng JL, Cook AL, Talbot J, Perry S. How is Excitotoxicity Being Modelled in iPSC-Derived Neurons? Neurotox Res 2024; 42:43. [PMID: 39405005 PMCID: PMC11480214 DOI: 10.1007/s12640-024-00721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Excitotoxicity linked either to environmental causes (pesticide and cyanotoxin exposure), excitatory neurotransmitter imbalance, or to intrinsic neuronal hyperexcitability, is a pathological mechanism central to neurodegeneration in amyotrophic lateral sclerosis (ALS). Investigation of excitotoxic mechanisms using in vitro and in vivo animal models has been central to understanding ALS mechanisms of disease. In particular, advances in induced pluripotent stem cell (iPSC) technologies now provide human cell-based models that are readily amenable to environmental and network-based excitotoxic manipulations. The cell-type specific differentiation of iPSC, combined with approaches to modelling excitotoxicity that include editing of disease-associated gene variants, chemogenetics, and environmental risk-associated exposures make iPSC primed to examine gene-environment interactions and disease-associated excitotoxic mechanisms. Critical to this is knowledge of which neurotransmitter receptor subunits are expressed by iPSC-derived neuronal cultures being studied, how their activity responds to antagonists and agonists of these receptors, and how to interpret data derived from multi-parameter electrophysiological recordings. This review explores how iPSC-based studies have contributed to our understanding of ALS-linked excitotoxicity and highlights novel approaches to inducing excitotoxicity in iPSC-derived neurons to further our understanding of its pathological pathways.
Collapse
Affiliation(s)
- Jan L Cheng
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Anthony L Cook
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Jana Talbot
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia.
| |
Collapse
|
20
|
Mirceta M, Schmidt MM, Shum N, Prasolava T, Meikle B, Lanni S, Mohiuddin M, McKeever P, Zhang M, Liang M, van der Werf I, Scheers S, Dion P, Wang P, Wilson M, Abell T, Philips E, Sznajder Ł, Swanson M, Mehkary M, Khan M, Yokoi K, Jung C, de Jong P, Freudenreich C, McGoldrick P, Yuen RC, Abrahão A, Keith J, Zinman L, Robertson J, Rogaeva E, Rouleau G, Kooy R, Pearson C. C9orf72 repeat expansion creates the unstable folate-sensitive fragile site FRA9A. NAR MOLECULAR MEDICINE 2024; 1:ugae019. [PMID: 39669124 PMCID: PMC11632612 DOI: 10.1093/narmme/ugae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024]
Abstract
The hyper-unstable Chr9p21 locus, harbouring the interferon gene cluster, oncogenes and C9orf72, is linked to multiple diseases. C9orf72 (GGGGCC)n expansions (C9orf72Exp) are associated with incompletely penetrant amyotrophic lateral sclerosis, frontotemporal dementia and autoimmune disorders. C9orf72Exp patients display hyperactive cGAS-STING-linked interferon immune and DNA damage responses, but the source of immunostimulatory or damaged DNA is unknown. Here, we show C9orf72Exp in pre-symptomatic and amyotrophic lateral sclerosis-frontotemporal dementia patient cells and brains cause the folate-sensitive chromosomal fragile site, FRA9A. FRA9A centers on >33 kb of C9orf72 as highly compacted chromatin embedded in an 8.2 Mb fragility zone spanning 9p21, encompassing 46 genes, making FRA9A one of the largest fragile sites. C9orf72Exp cells show chromosomal instability, heightened global- and Chr9p-enriched sister-chromatid exchanges, truncated-Chr9s, acentric-Chr9s and Chr9-containing micronuclei, providing endogenous sources of damaged and immunostimulatory DNA. Cells from one C9orf72Exp patient contained a highly rearranged FRA9A-expressing Chr9 with Chr9-wide dysregulated gene expression. Somatic C9orf72Exp repeat instability and chromosomal fragility are sensitive to folate deficiency. Age-dependent repeat instability, chromosomal fragility and chromosomal instability can be transferred to CNS and peripheral tissues of transgenic C9orf72Exp mice, implicating C9orf72Exp as the source. Our results highlight unappreciated effects of C9orf72 expansions that trigger vitamin-sensitive chromosome fragility, adding structural variations to the disease-enriched 9p21 locus, and likely elsewhere.
Collapse
Affiliation(s)
- Mila Mirceta
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Monika H M Schmidt
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Natalie Shum
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Tanya K Prasolava
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Bryanna Meikle
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Stella Lanni
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Mohiuddin Mohiuddin
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Paul M McKeever
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Ming Zhang
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, 200090, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Advanced Study, Tongji University, Shanghai, 200092, China
| | - Minggao Liang
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | | | - Stefaan Scheers
- Department of Medical Genetics, University of Antwerp, Belgium
| | - Patrick A Dion
- Montreal Neurological Institute-Hospital, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
| | - Peixiang Wang
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Michael D Wilson
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Theresa Abell
- Department of Biology, Tufts University, 200 Boston Avenue, Medford, MA 02155, USA
| | - Elliot A Philips
- Department of Biology, Tufts University, 200 Boston Avenue, Medford, MA 02155, USA
| | - Łukasz J Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, 2033 Mowry Road, Gainesville, FL 32610-3610, USA
- Department of Chemistry and Biochemistry, University of Nevada, 4003-4505 South Maryland Parkway, Las Vegas, NV 89154, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, 2033 Mowry Road, Gainesville, FL 32610-3610, USA
| | - Mustafa Mehkary
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Mahreen Khan
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Katsuyuki Yokoi
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
| | - Christine Jung
- BACPAC Resource Center, Children’s Hospital Oakland Research Institute, 25129 NE 42nd Pl, Redmond, WA 98053, USA
| | - Pieter J de Jong
- BACPAC Resource Center, Children’s Hospital Oakland Research Institute, 25129 NE 42nd Pl, Redmond, WA 98053, USA
| | | | - Philip McGoldrick
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Ryan K C Yuen
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| | - Agessandro Abrahão
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, North York, Toronto, ON, M4N 3M5, Canada
| | - Julia Keith
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, North York, Toronto, ON, M4N 3M5, Canada
| | - Lorne Zinman
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, North York, Toronto, ON, M4N 3M5, Canada
| | - Janice Robertson
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research of Neurodegenerative Diseases, University of Toronto, 60 Leonard Avenue, Toronto, M5T 2S8, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute-Hospital, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
- Department of Human Genetics, McGill University, 3801 University Avenue, Montreal, Quebec, H3A 2B4, Canada
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Belgium
| | - Christopher E Pearson
- Program of Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M3S 1A8, Canada
| |
Collapse
|
21
|
Arróspide Elgarresta M, Gerovska D, Soto-Gordoa M, Jauregui García ML, Merino Hernández ML, Araúzo-Bravo MJ. Chronic disease incidence explained by stepwise models and co-occurrence among them. iScience 2024; 27:110816. [PMID: 39290836 PMCID: PMC11407032 DOI: 10.1016/j.isci.2024.110816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/13/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Multimorbidity (MM) is the co-occurrence of two or more chronic diseases. We provided a dynamic approach revealing the MM complexity constructing a multistep incidence-age model for all patients with MM between 2014 and 2021 in the Basque Health System, Spain. The multistep model, with eight steps for males and nine for females, is a very well-fitting representation of MM. To gain insight into the MM components, we modeled the 19 diseases used to calculate the Charlson Comorbidity Index (CCI). We observed that the CCI diseases formed a complex interaction network. Hierarchical clustering of the incidence-age profiles clustered the CCI diseases into low- and high-risk of dying pathologies. Diseases with a higher number of steps are better represented by a multistep model. Anatomically, diseases associated with the central nervous system have the highest number of steps, followed by those associated with the kidney, heart, peripheral vasulature, pancreas, joints, cerebral vasculature, lung, stomach, and liver.
Collapse
Affiliation(s)
- Mikel Arróspide Elgarresta
- Computational Biology and Systems Biomedicine, Biogipuzkoa Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biogipuzkoa Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain
| | - Myrian Soto-Gordoa
- Biogipuzkoa Health Research Institute, San Sebastian-Donostia, Spain
- Mondragon University, Faculty of Engineering, Mondragon, Spain
| | - María L Jauregui García
- Biogipuzkoa Health Research Institute, San Sebastian-Donostia, Spain
- Tolosaldea Integrated Health Care Organization, Tolosa, Spain
| | - Marisa L Merino Hernández
- Biogipuzkoa Health Research Institute, San Sebastian-Donostia, Spain
- Bidasoa Integrated Health Care Organization, Hondarribia, Spain
- Research Network on Chronicity, Primary Care and Prevention and Health Promotion (RICAAPS), Kronikgune Group, Barakaldo, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biogipuzkoa Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain
- Basque Foundation for Science, IKERBASQUE, Calle María Díaz Harokoa 3, 48013 Bilbao, Spain
- CIBER of Frailty and Healthy Aging (CIBERfes), 28029 Madrid, Spain
- Max Planck Institute for Molecular Biomedicine, Computational Biology and Bioinformatics, Röntgenstr. 20, 48149 Münster, Germany
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain
| |
Collapse
|
22
|
Feng H, Li J, Wang H, Wei Z, Feng S. Senescence- and Immunity-Related Changes in the Central Nervous System: A Comprehensive Review. Aging Dis 2024:AD.2024.0755. [PMID: 39325939 DOI: 10.14336/ad.2024.0755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Senescence is a cellular state characterized by an irreversible halt in the cell cycle, accompanied by alterations in cell morphology, function, and secretion. Senescent cells release a plethora of inflammatory and growth factors, extracellular matrix proteins, and other bioactive substances, collectively known as the senescence-associated secretory phenotype (SASP). These excreted substances serve as crucial mediators of senescent tissues, while the secretion of SASP by senescent neurons and glial cells in the central nervous system modulates the activity of immune cells. Senescent immune cells also influence the physiological activities of various cells in the central nervous system. Further, the interaction between cellular senescence and immune regulation collectively affects the physiological and pathological processes of the central nervous system. Herein, we explore the role of senescence in the physiological and pathological processes underlying embryonic development, aging, degeneration, and injury of the central nervous system, through the immune response. Further, we elucidate the role of senescence in the physiological and pathological processes of the central nervous system, proposing a new theoretical foundation for treating central nervous system diseases.
Collapse
Affiliation(s)
- Haiwen Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Junjin Li
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Hongda Wang
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Zhijian Wei
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shiqing Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
23
|
Chourpiliadis C, Seitz C, Lovik A, Joyce EE, Pan L, Hu Y, Kläppe U, Samuelsson K, Press R, Ingre C, Fang F. Lifestyle and medical conditions in relation to ALS risk and progression-an introduction to the Swedish ALSrisc Study. J Neurol 2024; 271:5447-5459. [PMID: 38878106 PMCID: PMC11319377 DOI: 10.1007/s00415-024-12496-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND This study was an introduction to the Swedish ALSrisc Study and explored the association of lifestyle and medical conditions, with risk and progression of amyotrophic lateral sclerosis (ALS). METHODS We included 265 newly diagnosed ALS patients during 2016-2022 in Stockholm and 207 ALS-free siblings and partners of the patients as controls. Information on body mass index (BMI), smoking, and history of head injuries, diabetes mellitus, hypercholesterolemia, and hypertension was obtained through the Euro-MOTOR questionnaire at recruitment. Patients were followed from diagnosis until death, invasive ventilation, or November 30, 2022. RESULTS Higher BMI at recruitment was associated with lower risk for ALS (OR 0.89, 95%CI 0.83-0.95), especially among those diagnosed after 65 years. One unit increase in the average BMI during the 3 decades before diagnosis was associated with a lower risk for ALS (OR 0.94, 95%CI 0.89-0.99). Diabetes was associated with lower risk of ALS (OR 0.38, 95%CI 0.16-0.90), while hypercholesterolemia was associated with higher risk of ALS (OR 2.10, 95%CI 1.13-3.90). Higher BMI at diagnosis was associated with lower risk of death (HR 0.91, 95%CI 0.84-0.98), while the highest level of smoking exposure (in pack-years) (HR 1.90, 95%CI 1.20-3.00), hypercholesterolemia (HR 1.84, 95%CI 1.06-3.19), and hypertension (HR 1.76, 95%CI 1.03-3.01) were associated with higher risk of death, following ALS diagnosis. CONCLUSIONS Higher BMI and diabetes were associated with lower risk of ALS. Higher BMI was associated with lower risk of death, whereas smoking (especially in high pack-years), hypercholesterolemia, and hypertension were associated with higher risk of death after ALS diagnosis.
Collapse
Affiliation(s)
| | - Christina Seitz
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anikó Lovik
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Institute of Psychology, Leiden University, Leiden, The Netherlands
| | - Emily E Joyce
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lu Pan
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yihan Hu
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Kläppe
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Kristin Samuelsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Rayomand Press
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Ingre
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Leighton DJ, Ansari M, Newton J, Cleary E, Stephenson L, Beswick E, Carod Artal J, Davenport R, Duncan C, Gorrie GH, Morrison I, Swingler R, Deary IJ, Porteous M, Chandran S, Pal S. Genotypes and phenotypes of motor neuron disease: an update of the genetic landscape in Scotland. J Neurol 2024; 271:5256-5266. [PMID: 38852112 PMCID: PMC11319561 DOI: 10.1007/s00415-024-12450-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/09/2024] [Accepted: 05/16/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Using the Clinical Audit Research and Evaluation of Motor Neuron Disease (CARE-MND) database and the Scottish Regenerative Neurology Tissue Bank, we aimed to outline the genetic epidemiology and phenotypes of an incident cohort of people with MND (pwMND) to gain a realistic impression of the genetic landscape and genotype-phenotype associations. METHODS Phenotypic markers were identified from the CARE-MND platform. Sequence analysis of 48 genes was undertaken. Variants were classified using a structured evidence-based approach. Samples were also tested for C9orf72 hexanucleotide expansions using repeat-prime PCR methodology. RESULTS 339 pwMND donated a DNA sample: 44 (13.0%) fulfilled criteria for having a pathogenic variant/repeat expansion, 53.5% of those with a family history of MND and 9.3% of those without. The majority (30 (8.8%)) had a pathogenic C9orf72 repeat expansion, including two with intermediate expansions. Having a C9orf72 expansion was associated with a significantly lower Edinburgh Cognitive and Behavioural ALS Screen ALS-Specific score (p = 0.0005). The known pathogenic SOD1 variant p.(Ile114Thr), frequently observed in the Scottish population, was detected in 9 (2.7%) of total cases but in 17.9% of familial cases. Rare variants were detected in FUS and NEK1. One individual carried both a C9orf72 expansion and SOD1 variant. CONCLUSIONS Our results provide an accurate summary of MND demographics and genetic epidemiology. We recommend early genetic testing of people with cognitive impairment to ensure that C9orf72 carriers are given the best opportunity for informed treatment planning. Scotland is enriched for the SOD1 p.(Ile114Thr) variant and this has significant implications with regards to future genetically-targeted treatments.
Collapse
Affiliation(s)
- Danielle J Leighton
- School of Psychology & Neuroscience, University of Glasgow, Glasgow, UK.
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK.
- Anne Rowling Regenerative Neurology Clinic, Royal Infirmary, Edinburgh, UK.
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Institute of Neurological Sciences, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK.
| | - Morad Ansari
- South East Scotland Genetics Service, Western General Hospital, Edinburgh, UK
| | - Judith Newton
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, Royal Infirmary, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Elaine Cleary
- South East Scotland Genetics Service, Western General Hospital, Edinburgh, UK
| | - Laura Stephenson
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Emily Beswick
- Anne Rowling Regenerative Neurology Clinic, Royal Infirmary, Edinburgh, UK
| | | | - Richard Davenport
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, Royal Infirmary, Edinburgh, UK
| | - Callum Duncan
- Department of Neurology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - George H Gorrie
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
- Institute of Neurological Sciences, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK
| | - Ian Morrison
- Department of Neurology, NHS Tayside, Dundee, UK
| | - Robert Swingler
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Lothian Birth Cohorts Group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Mary Porteous
- South East Scotland Genetics Service, Western General Hospital, Edinburgh, UK
| | - Siddharthan Chandran
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, Royal Infirmary, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Suvankar Pal
- The Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, Royal Infirmary, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
25
|
Ciuro M, Sangiorgio M, Cacciato V, Cantone G, Fichera C, Salvatorelli L, Magro G, Leanza G, Vecchio M, Valle MS, Gulino R. Mitigating the Functional Deficit after Neurotoxic Motoneuronal Loss by an Inhibitor of Mitochondrial Fission. Int J Mol Sci 2024; 25:7059. [PMID: 39000168 PMCID: PMC11241433 DOI: 10.3390/ijms25137059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an extremely complex neurodegenerative disease involving different cell types, but motoneuronal loss represents its main pathological feature. Moreover, compensatory plastic changes taking place in parallel to neurodegeneration are likely to affect the timing of ALS onset and progression and, interestingly, they might represent a promising target for disease-modifying treatments. Therefore, a simplified animal model mimicking motoneuronal loss without the other pathological aspects of ALS has been established by means of intramuscular injection of cholera toxin-B saporin (CTB-Sap), which is a targeted neurotoxin able to kill motoneurons by retrograde suicide transport. Previous studies employing the mouse CTB-Sap model have proven that spontaneous motor recovery is possible after a subtotal removal of a spinal motoneuronal pool. Although these kinds of plastic changes are not enough to counteract the functional effects of the progressive motoneuron degeneration, it would nevertheless represent a promising target for treatments aiming to postpone ALS onset and/or delay disease progression. Herein, the mouse CTB-Sap model has been used to test the efficacy of mitochondrial division inhibitor 1 (Mdivi-1) as a tool to counteract the CTB-Sap toxicity and/or to promote neuroplasticity. The homeostasis of mitochondrial fission/fusion dynamics is indeed important for cell integrity, and it could be affected during neurodegeneration. Lesioned mice were treated with Mdivi-1 and then examined by a series of behavioral test and histological analyses. The results have shown that the drug may be capable of reducing functional deficits after the lesion and promoting synaptic plasticity and neuroprotection, thus representing a putative translational approach for motoneuron disorders.
Collapse
Affiliation(s)
- Maria Ciuro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| | - Maria Sangiorgio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| | - Valeria Cacciato
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| | - Giuliano Cantone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| | - Carlo Fichera
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| | - Lucia Salvatorelli
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (L.S.); (G.M.)
| | - Gaetano Magro
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (L.S.); (G.M.)
| | - Giampiero Leanza
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
| | - Michele Vecchio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| | - Maria Stella Valle
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.); (M.S.); (V.C.); (G.C.); (C.F.); (M.V.); (M.S.V.)
| |
Collapse
|
26
|
Romer SH, Miller KM, Sonner MJ, Ethridge VT, Gargas NM, Rohan JG. Changes in motor behavior and lumbar motoneuron morphology following repeated chlorpyrifos exposure in rats. PLoS One 2024; 19:e0305173. [PMID: 38875300 PMCID: PMC11178230 DOI: 10.1371/journal.pone.0305173] [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: 02/23/2024] [Accepted: 05/26/2024] [Indexed: 06/16/2024] Open
Abstract
Chlorpyrifos is an organophosphate pesticide associated with numerous health effects including motor performance decrements. While many studies have focused on the health effects following acute chlorpyrifos poisonings, almost no studies have examined the effects on motoneurons following occupational-like exposures. The main objective of this study was to examine the broad effects of repeated occupational-like chlorpyrifos exposures on spinal motoneuron soma size relative to motor activity. To execute our objective, adult rats were exposed to chlorpyrifos via oral gavage once a day, five days a week for two weeks. Chlorpyrifos exposure effects were assessed either three days or two months following the last exposure. Three days following the last repeated chlorpyrifos exposure, there were transient effects in open-field motor activity and plasma cholinesterase activity levels. Two months following the chlorpyrifos exposures, there were delayed effects in sensorimotor gating, pro-inflammatory cytokines and spinal lumbar motoneuron soma morphology. Overall, these results offer support that subacute repeated occupational-like chlorpyrifos exposures have both short-term and longer-term effects in motor activity, inflammation, and central nervous system mechanisms.
Collapse
Affiliation(s)
- Shannon H Romer
- Environmental Health Effects Laboratory, Naval Medical Research Unit Dayton, Wright-Patterson AFB, Dayton, OH, United States of America
- Leidos, Reston, VA, United States of America
| | - Kaitlyn M Miller
- Environmental Health Effects Laboratory, Naval Medical Research Unit Dayton, Wright-Patterson AFB, Dayton, OH, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, United States of America
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, United States of America
| | - Martha J Sonner
- Environmental Health Effects Laboratory, Naval Medical Research Unit Dayton, Wright-Patterson AFB, Dayton, OH, United States of America
- Leidos, Reston, VA, United States of America
| | - Victoria T Ethridge
- Environmental Health Effects Laboratory, Naval Medical Research Unit Dayton, Wright-Patterson AFB, Dayton, OH, United States of America
- Leidos, Reston, VA, United States of America
| | - Nathan M Gargas
- Environmental Health Effects Laboratory, Naval Medical Research Unit Dayton, Wright-Patterson AFB, Dayton, OH, United States of America
| | - Joyce G Rohan
- Environmental Health Effects Laboratory, Naval Medical Research Unit Dayton, Wright-Patterson AFB, Dayton, OH, United States of America
| |
Collapse
|
27
|
Al-Chalabi A, Andrews J, Farhan S. Recent advances in the genetics of familial and sporadic ALS. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:49-74. [PMID: 38802182 DOI: 10.1016/bs.irn.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
ALS shows complex genetic inheritance patterns. In about 5% to 10% of cases, there is a family history of ALS or a related condition such as frontotemporal dementia in a first or second degree relative, and for about 80% of such people a pathogenic gene variant can be identified. Such variants are also seen in people with no family history because of factor influencing the expression of genes, such as age. Genetic susceptibility factors also contribute to risk, and the heritability of ALS is between 40% and 60%. The genetic variants influencing ALS risk include single base changes, repeat expansions, copy number variants, and others. Here we review what is known of the genetic landscape and architecture of ALS.
Collapse
Affiliation(s)
- Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, King's College London, London, United Kingdom.
| | - Jinsy Andrews
- Department of Neurology, Columbia University, New York, NY, United States
| | - Sali Farhan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Montreal, QC, Canada; Department of Human Genetics, Montreal Neurological Institute-Hospital, Montreal, QC, Canada
| |
Collapse
|
28
|
Pandya VA, Patani R. The role of glial cells in amyotrophic lateral sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:381-450. [PMID: 38802179 DOI: 10.1016/bs.irn.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) has traditionally been considered a neuron-centric disease. This view is now outdated, with increasing recognition of cell autonomous and non-cell autonomous contributions of central and peripheral nervous system glia to ALS pathomechanisms. With glial research rapidly accelerating, we comprehensively interrogate the roles of astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells and satellite glia in nervous system physiology and ALS-associated pathology. Moreover, we highlight the inter-glial, glial-neuronal and inter-system polylogue which constitutes the healthy nervous system and destabilises in disease. We also propose classification based on function for complex glial reactive phenotypes and discuss the pre-requisite for integrative modelling to advance translation. Given the paucity of life-enhancing therapies currently available for ALS patients, we discuss the promising potential of harnessing glia in driving ALS therapeutic discovery.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- University College London Medical School, London, United Kingdom; The Francis Crick Institute, London, United Kingdom.
| | - Rickie Patani
- The Francis Crick Institute, London, United Kingdom; Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, United Kingdom.
| |
Collapse
|
29
|
Moll T, Harvey C, Alhathli E, Gornall S, O'Brien D, Cooper-Knock J. Non-coding genome contribution to ALS. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:75-86. [PMID: 38802183 DOI: 10.1016/bs.irn.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The majority of amyotrophic lateral sclerosis (ALS) is caused by a complex gene-environment interaction. Despite high estimates of heritability, the genetic basis of disease in the majority of ALS patients are unknown. This limits the development of targeted genetic therapies which require an understanding of patient-specific genetic drivers. There is good evidence that the majority of these missing genetic risk factors are likely to be found within the non-coding genome. However, a major challenge in the discovery of non-coding risk variants is determining which variants are functional in which specific CNS cell type. We summarise current discoveries of ALS-associated genetic drivers within the non-coding genome and we make the case that improved cell-specific annotation of genomic function is required to advance this field, particularly via single-cell epigenetic profiling and spatial transcriptomics. We highlight the example of TBK1 where an apparent paradox exists between pathogenic coding variants which cause loss of protein function, and protective non-coding variants which cause reduced gene expression; the paradox is resolved when it is understood that the non-coding variants are acting primarily via change in gene expression within microglia, and the effect of coding variants is most prominent in neurons. We propose that cell-specific functional annotation of ALS-associated genetic variants will accelerate discovery of the genetic architecture underpinning disease in the vast majority of patients.
Collapse
Affiliation(s)
- Tobias Moll
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Calum Harvey
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Elham Alhathli
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Sarah Gornall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - David O'Brien
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
30
|
Shelkovnikova TA, Hautbergue GM. RNP granules in ALS and neurodegeneration: From multifunctional membraneless organelles to therapeutic opportunities. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:455-479. [PMID: 38802180 DOI: 10.1016/bs.irn.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) and related neurodegenerative diseases are characterised by dysfunction of a host of RNA-binding proteins (RBPs) and a severely disrupted RNA metabolism. Recently, RBP-harbouring phase-separated complexes, ribonucleoprotein (RNP) granules, have come into the limelight as "crucibles" of neuronal pathology in ALS. RNP granules are indispensable for the multitude of regulatory processes underlying cellular RNA metabolism and serve as critical organisers of cellular biochemistry. Neurons, highly specialised cells, heavily rely on RNP granules for efficient trafficking, signalling and stress responses. Multiple RNP granule components, primarily RBPs such as TDP-43 and FUS, are affected by ALS mutations. However, even in the absence of mutations, RBP proteinopathies represent pathophysiological hallmarks of ALS. Given the high local concentrations of RBPs and RNAs, their weakened or enhanced interactions within RNP granules disrupt their homeostasis. Thus, the physiological process of phase separation and RNP granule formation, vital for maintaining the high-functioning state of neuronal cells, becomes their Achilles heel. Here, we will review the recent literature on the causes and consequences of abnormal RNP granule functioning in ALS and related disorders. In particular, we will summarise the evidence for the network-level dysfunction of RNP granules in these conditions and discuss considerations for therapeutic interventions to target RBPs, RNP granules and their network as a whole.
Collapse
Affiliation(s)
- Tatyana A Shelkovnikova
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom; Neuroscience Institute, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom.
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom; Neuroscience Institute, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom; Healthy Lifespan Institute (HELSI), University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom.
| |
Collapse
|
31
|
Pokrishevsky E, DuVal MG, McAlary L, Louadi S, Pozzi S, Roman A, Plotkin SS, Dijkstra A, Julien JP, Allison WT, Cashman NR. Tryptophan residues in TDP-43 and SOD1 modulate the cross-seeding and toxicity of SOD1. J Biol Chem 2024; 300:107207. [PMID: 38522514 PMCID: PMC11087967 DOI: 10.1016/j.jbc.2024.107207] [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: 10/02/2023] [Revised: 02/04/2024] [Accepted: 03/05/2024] [Indexed: 03/26/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of motor neurons. Neuronal superoxide dismutase-1 (SOD1) inclusion bodies are characteristic of familial ALS with SOD1 mutations, while a hallmark of sporadic ALS is inclusions containing aggregated WT TAR DNA-binding protein 43 (TDP-43). We show here that co-expression of mutant or WT TDP-43 with SOD1 leads to misfolding of endogenous SOD1 and aggregation of SOD1 reporter protein SOD1G85R-GFP in human cell cultures and promotes synergistic axonopathy in zebrafish. Intriguingly, this pathological interaction is modulated by natively solvent-exposed tryptophans in SOD1 (tryptophan-32) and TDP-43 RNA-recognition motif RRM1 (tryptophan-172), in concert with natively sequestered TDP-43 N-terminal domain tryptophan-68. TDP-43 RRM1 intrabodies reduce WT SOD1 misfolding in human cell cultures, via blocking tryptophan-172. Tryptophan-68 becomes antibody-accessible in aggregated TDP-43 in sporadic ALS motor neurons and cell culture. 5-fluorouridine inhibits TDP-43-induced G85R-GFP SOD1 aggregation in human cell cultures and ameliorates axonopathy in zebrafish, via its interaction with SOD1 tryptophan-32. Collectively, our results establish a novel and potentially druggable tryptophan-mediated mechanism whereby two principal ALS disease effector proteins might directly interact in disease.
Collapse
Affiliation(s)
- Edward Pokrishevsky
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michéle G DuVal
- Department of Biological Sciences, Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Alberta, Canada
| | - Luke McAlary
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah Louadi
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Silvia Pozzi
- Department of Psychiatry and Neuroscience, University of Laval, Québec, Quebec, Canada; CERVO Brain Research Center, Québec, Quebec, Canada
| | - Andrei Roman
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven S Plotkin
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anke Dijkstra
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, University of Laval, Québec, Quebec, Canada; CERVO Brain Research Center, Québec, Quebec, Canada
| | - W Ted Allison
- Department of Biological Sciences, Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Alberta, Canada.
| | - Neil R Cashman
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
32
|
Stenson K, Fecteau TE, O'Callaghan L, Bryden P, Mellor J, Wright J, Earl L, Thomas O, Iqbal H, Barlow S, Parvanta S. Health-related quality of life across disease stages in patients with amyotrophic lateral sclerosis: results from a real-world survey. J Neurol 2024; 271:2390-2404. [PMID: 38200398 PMCID: PMC11055770 DOI: 10.1007/s00415-023-12141-y] [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: 08/24/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is characterized by a rapid disease course, with disease severity being associated with declining health-related quality of life (HRQoL) in persons living with ALS (pALS). The main objective of this study was to assess the impact of disease progression on HRQoL across King's, Milano-Torino Staging (MiToS), and physician-judgement clinical staging. Additionally, we evaluated the impact of the disease on the HRQoL of care partners (cALS). METHODS Data were sourced from the Adelphi ALS Disease Specific Programme (DSP)™, a cross-sectional survey of neurologists, pALS and cALS presenting in a real-world clinical setting between July 2020 and March 2021 in Europe and the United States. RESULTS Neurologists (n = 142) provided data for 880 pALS. There were significant negative correlations between all three clinical staging systems and EuroQol (European Quality of Life) Five Dimension Five Level Scale (EQ-5D-5L) utility scores and visual analogue scale (VAS) ratings. Although not all differences were significant, 5-item Amyotrophic Lateral Sclerosis Assessment Questionnaire (ALSAQ-5) scores showed a stepwise increase in HRQoL impairment at each stage of the disease regardless of the staging system. At later stages, high levels of fatigue and substantial activity impairment were reported. As pALS disease states progressed, cALS also experienced a decline in HRQoL and increased burden. CONCLUSIONS Across outcomes, pALS and cALS generally reported worse outcomes at later stages of the disease, highlighting an unmet need in this population for strategies to maximise QoL despite disease progression. Recognition and treatment of symptoms such as pain and fatigue may lead to improved outcomes for pALS and cALS.
Collapse
Affiliation(s)
| | | | - L O'Callaghan
- Biogen, Cambridge, MA, USA
- Sage Therapeutics, Boston, MA, USA
| | | | - J Mellor
- Adelphi Real World, Bollington, UK
| | - J Wright
- Adelphi Real World, Bollington, UK
| | - L Earl
- Adelphi Real World, Bollington, UK
| | - O Thomas
- Adelphi Real World, Bollington, UK
| | - H Iqbal
- Adelphi Real World, Bollington, UK
| | - S Barlow
- Adelphi Real World, Bollington, UK
| | | |
Collapse
|
33
|
Ferguson R, van Es MA, van den Berg LH, Subramanian V. Neural stem cell homeostasis is affected in cortical organoids carrying a mutation in Angiogenin. J Pathol 2024; 262:410-426. [PMID: 38180358 DOI: 10.1002/path.6244] [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: 04/12/2023] [Revised: 11/07/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
Mutations in Angiogenin (ANG) and TARDBP encoding the 43 kDa transactive response DNA binding protein (TDP-43) are associated with amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD). ANG is neuroprotective and plays a role in stem cell dynamics in the haematopoietic system. We obtained skin fibroblasts from members of an ALS-FTD family, one with mutation in ANG, one with mutation in both TARDBP and ANG, and one with neither mutation. We reprogrammed these fibroblasts to induced pluripotent stem cells (iPSCs) and generated cortical organoids as well as induced stage-wise differentiation of the iPSCs to neurons. Using these two approaches we investigated the effects of FTD-associated mutations in ANG and TARDBP on neural precursor cells, neural differentiation, and response to stress. We observed striking neurodevelopmental defects such as abnormal and persistent rosettes in the organoids accompanied by increased self-renewal of neural precursor cells. There was also a propensity for differentiation to later-born neurons. In addition, cortical neurons showed increased susceptibility to stress, which is exacerbated in neurons carrying mutations in both ANG and TARDBP. The cortical organoids and neurons generated from patient-derived iPSCs carrying ANG and TARDBP gene variants recapitulate dysfunctions characteristic of frontotemporal lobar degeneration observed in FTD patients. These dysfunctions were ameliorated upon treatment with wild type ANG. In addition to its well-established role during the stress response of mature neurons, ANG also appears to play a role in neural progenitor dynamics. This has implications for neurogenesis and may indicate that subtle developmental defects play a role in disease susceptibility or onset. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ross Ferguson
- Department of Life Sciences, University of Bath, Bath, UK
| | - Michael A van Es
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
34
|
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
|
35
|
Thompson AG, Taschler B, Smith SM, Turner MR. Premorbid brain structure influences risk of amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2024; 95:360-365. [PMID: 38050140 PMCID: PMC10958375 DOI: 10.1136/jnnp-2023-332322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a disease of the motor network associated with brain structure and functional connectivity alterations that are implicated in disease progression. Whether such changes have a causal role in ALS, fitting with a postulated influence of premorbid cerebral architecture on the phenotypes associated with neurodegenerative disorders is not known. METHODS This study considered causal effects and shared genetic risk of 2240 structural and functional MRI brain scan imaging-derived phenotypes (IDPs) on ALS using two sample Mendelian randomisation, with putative associations further examined with extensive sensitivity analysis. Shared genetic predisposition between IDPs and ALS was explored using genetic correlation analysis. RESULTS Increased white matter volume in the cerebral hemispheres was causally associated with ALS. Weaker causal associations were observed for brain stem grey matter volume, parieto-occipital white matter surface and volume of the left thalamic ventral anterior nucleus. Genetic correlation was observed between ALS and intracellular volume fraction and isotropic free water volume fraction within the posterior limb of the internal capsule. CONCLUSIONS This study provides evidence that premorbid brain structure, in particular white matter volume, contributes to the risk of ALS.
Collapse
Affiliation(s)
| | - Bernd Taschler
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Stephen M Smith
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Holdom CJ, Janse van Mantgem MR, He J, Howe SL, McCombe PA, Fan D, van den Berg LH, Henderson RD, van Eijk R, Steyn FJ, Ngo ST. Variation in Resting Metabolic Rate Affects Identification of Metabolic Change in Geographically Distinct Cohorts of Patients With ALS. Neurology 2024; 102:e208117. [PMID: 38350046 DOI: 10.1212/wnl.0000000000208117] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/16/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Altered metabolism is observed in amyotrophic lateral sclerosis (ALS). However, without a standardized methodology to define metabolic changes, our understanding of factors contributing to and the clinical significance of altered metabolism in ALS is limited. METHODS We aimed to determine how geographic variation in metabolic rates influences estimates and accuracy of predicted resting energy expenditure (REE) in patients with ALS and controls, while validating the effectiveness of cohort-specific approaches in predicting altered metabolic rate in ALS. Participants from 3 geographically distinct sites across Australia, China, and the Netherlands underwent REE assessments, and we considered 22 unique equations for estimating REE. Analyses evaluated equation performance and the influence of demographics on metabolic status. Comparisons were made using standardized and local reference values to identify metabolic alterations. RESULTS 606 participants were included from Australia (patients with ALS: 140, controls: 154), the Netherlands (patients with ALS: 79, controls: 37) and China (patients with ALS: 67, controls: 129). Measured REE was variable across geographic cohorts, with fat-free mass contributing to this variation across all patients (p = 0.002 to p < 0.001). Of the 22 predication equations assessed, the Sabounchi Structure 4 (S4) equation performed relatively well across all control cohorts. Use of prediction thresholds generated using data from Australian controls generally increased the prevalence of hypermetabolism in Chinese (55%, [43%-67%]) and Dutch (44%, [33%-55%]) cases when compared with Australian cases (30%, [22%-38%]). Adjustment of prediction thresholds to consider geographically distinct characteristics from matched control cohorts resulted in a decrease in the proportion of hypermetabolic cases in Chinese and Dutch cohorts (25%-31% vs 55% and 20%-34% vs 43%-44%, respectively), and increased prevalence of hypometabolism in Dutch cases with ALS (1% to 8%-10%). DISCUSSION The identification of hypermetabolism in ALS is influenced by the formulae and demographic-specific prediction thresholds used for defining alterations in metabolic rate. A consensus approach is needed for identification of metabolic changes in ALS and will facilitate improved understanding of the cause and clinical significance of this in ALS.
Collapse
Affiliation(s)
- Cory J Holdom
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Mark R Janse van Mantgem
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Ji He
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Stephanie L Howe
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Pamela A McCombe
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Dongsheng Fan
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Leonard H van den Berg
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Robert D Henderson
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Ruben van Eijk
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Frederik J Steyn
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| | - Shyuan T Ngo
- From the Australian Institute for Bioengineering and Nanotechnology (C.J.H., S.L.H., S.T.N.), The University of Queensland, Australia; Department of Neurology (M.R.J.M., R.P.A.E., L.H.B.), UMC Utrecht Brain Centre, University Medical Centre Utrecht, The Netherlands; Department of Neurology (D.F.), Peking University Third Hospital; Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases (D.F.), China; Centre for Clinical Research (P.A.M., R.D.H., F.J.S., S.T.N.), The University of Queensland; Department of Neurology (P.A.M., R.D.H., F.J.S., S.T.N.), Royal Brisbane and Women's Hospital, Australia; Biostatistics and Research Support (R.P.A.E.), Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, The Netherlands; and School of Biomedical Sciences (F.J.S.), The University of Queensland, Australia
| |
Collapse
|
37
|
Domi T, Schito P, Sferruzza G, Russo T, Pozzi L, Agosta F, Carrera P, Riva N, Filippi M, Quattrini A, Falzone YM. Unveiling the SOD1-mediated ALS phenotype: insights from a comprehensive meta-analysis. J Neurol 2024; 271:1342-1354. [PMID: 37930481 DOI: 10.1007/s00415-023-12074-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND AND OBJECTIVES Amyotrophic lateral sclerosis associated with mutations in SOD1 (SOD1-ALS) might be susceptible to specific treatment. The aim of the study is to outline the clinical features of SOD1-ALS patients by comparing them to patients without ALS major gene variants and patients with variants in other major ALS genes. Defining SOD1-ALS phenotype may assist clinicians in identifying patients who should be prioritized for genetic testing. METHODS We performed an extensive literature research including original studies which reported the clinical features of SOD1-ALS and at least one of the following patient groups: C9ORF72 hexanucleotide repeat expansion (C9-ALS), TARDBP (TARDBP-ALS), FUS (FUS-ALS) or patients without a positive test for a major-ALS gene (N-ALS). A random effects meta-analytic model was applied to clinical data extracted encompassing sex, site and age of onset. To reconstruct individual patient survival data, the published Kaplan-Meier curves were digitized. Data were measured as odds ratio (OR) or standardized mean difference (SMD) as appropriate. Median survival was compared between groups. RESULTS Twenty studies met the inclusion criteria. We identified 721 SOD1-ALS, 470 C9-ALS, 183 TARDBP-ALS, 113 FUS-ALS and 2824 N-ALS. SOD1-ALS showed a higher rate of spinal onset compared with N-ALS and C9-ALS (OR = 4.85, 95% CI = 3.04-7.76; OR = 10.47, 95% CI = 4.32-27.87) and an earlier onset compared with N-ALS (SMD = - 0.45, 95% CI = - 0.72 to - 0.18). SOD1-ALS had a similar survival compared with N-ALS (p = 0.14), a longer survival compared with C9-ALS (p < 0.01) and FUS-ALS (p = 0.019) and a shorter survival compared with TARDBP-ALS (p < 0.01). DISCUSSION This study indicates the presence of a specific SOD1-ALS phenotype. Insights in SOD1-ALS clinical features are important in genetic counseling, disease prognosis and support patients' stratification in clinical trials.
Collapse
Affiliation(s)
- Teuta Domi
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paride Schito
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Giacomo Sferruzza
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Tommaso Russo
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| | - Laura Pozzi
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Agosta
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroimaging Research Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis, Division of Genetics and Cell Biology, Laboratory of Clinical Molecular Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nilo Riva
- 3rd Neurology Unit and Motor Neuron Disease Centre, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Massimo Filippi
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
- Neuroimaging Research Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Angelo Quattrini
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Yuri Matteo Falzone
- Experimental Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| |
Collapse
|
38
|
Van Wijk IF, Van Eijk RPA, Van Boxmeer L, Westeneng HJ, Van Es MA, Van Rheenen W, Van Den Berg LH, Eijkemans MJC, Veldink JH. Assessment of risk of ALS conferred by the GGGGCC hexanucleotide repeat expansion in C9orf72 among first-degree relatives of patients with ALS carrying the repeat expansion. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:188-196. [PMID: 37861203 DOI: 10.1080/21678421.2023.2272187] [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: 07/26/2023] [Accepted: 10/08/2023] [Indexed: 10/21/2023]
Abstract
OBJECTIVES We aimed to estimate the age-related risk of ALS in first-degree relatives of patients with ALS carrying the C9orf72 repeat expansion. METHODS We included all patients with ALS carrying a C9orf72 repeat expansion in The Netherlands. Using structured questionnaires, we determined the number of first-degree relatives, their age at death due to ALS or another cause, or age at time of questionnaire. The cumulative incidence of ALS among first-degree relatives was estimated, while accounting for death from other causes. Variability in ALS risk between families was evaluated using a random effects hazards model. We used a second, distinct approach to estimate the risk of ALS and FTD in the general population, using previously published data. RESULTS In total, 214 of the 2,486 (9.2%) patients with ALS carried the C9orf72 repeat expansion. The mean risk of ALS at age 80 for first-degree relatives carrying the repeat expansion was 24.1%, but ranged between individual families from 16.0 to 60.6%. Using the second approach, we found the risk of ALS and FTD combined was 28.7% (95% CI 17.8%-54.3%) for carriers in the general population. CONCLUSIONS On average, our estimated risk of ALS in the C9orf72 repeat expansion was lower compared to historical estimates. We showed, however, that the risk of ALS likely varies between families and one overall penetrance estimate may not be sufficient to describe ALS risk. This warrants a tailor-made, patient-specific approach in testing. Further studies are needed to assess the risk of FTD in the C9orf72 repeat expansion.
Collapse
Affiliation(s)
- Iris F Van Wijk
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Ruben P A Van Eijk
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Loes Van Boxmeer
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Michael A Van Es
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Wouter Van Rheenen
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Leonard H Van Den Berg
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Marinus J C Eijkemans
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| | - Jan H Veldink
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands and
- Research support and Biostatistics, Julius Centre for Health Sciences and Primary Care, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
39
|
Ruigrok YM, Rinkel GJE, Chang H, Hackenberg KAM, Etminan N, Veldink JH. Analysis of aneurysmal subarachnoid hemorrhage as a multistep process. Eur J Neurol 2024; 31:e16118. [PMID: 37877684 PMCID: PMC11235647 DOI: 10.1111/ene.16118] [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: 08/18/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND AND PURPOSE Aneurysmal subarachnoid hemorrhage (ASAH) is a complex disease with higher incidence in women compared to men and in Japan compared to other countries. It was hypothesized that ASAH is consistent with a multistep model of disease. The following assessments were made: (1) the number of steps needed for the disease to occur and (2) whether this number may be different in female versus male and in Japanese versus non-Japanese patients. METHODS Incidence data were generated from a meta-analysis on ASAH incidence until 2017, which was supplemented with a literature search from 2017 to April 2023. Age- and sex-adjusted incidences per 10-year age groups were calculated and the logarithm of age-specific incidence against the logarithm of age was regressed with least-squares regression. RESULTS In 2317 ASAH patients a linear relationship between logarithm of incidence and logarithm of age was found with a slope estimate of 3.13 (95% confidence interval 2.60-3.65), consistent with a four-step process. Similar estimates were found for female, male, Japanese and non-Japanese patients. CONCLUSIONS Our results suggest that ASAH is a four-step process, also in subgroups with higher ASAH incidence. Elucidation of the exact nature of these steps can provide important clues for identification of disease mechanisms underlying ASAH.
Collapse
Affiliation(s)
- Ynte M. Ruigrok
- Department of NeurologyUniversity Medical Center Utrecht Brain Center, University Medical Center UtrechtUtrechtThe Netherlands
| | - Gabriel J. E. Rinkel
- Department of NeurologyUniversity Medical Center Utrecht Brain Center, University Medical Center UtrechtUtrechtThe Netherlands
| | - Han‐Sol Chang
- Department of NeurosurgeryMannheim University Hospital, Medical Faculty Mannheim, Heidelberg UniversityMannheimGermany
| | - Katharina A. M. Hackenberg
- Department of NeurosurgeryMannheim University Hospital, Medical Faculty Mannheim, Heidelberg UniversityMannheimGermany
| | - Nima Etminan
- Department of NeurosurgeryMannheim University Hospital, Medical Faculty Mannheim, Heidelberg UniversityMannheimGermany
| | - Jan H. Veldink
- Department of NeurologyUniversity Medical Center Utrecht Brain Center, University Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
40
|
Ryan M, Doherty MA, Al Khleifat A, Costello E, Hengeveld JC, Heverin M, Al-Chalabi A, Mclaughlin RL, Hardiman O. C9orf72 Repeat Expansion Discordance in 6 Multigenerational Kindreds. Neurol Genet 2024; 10:e200112. [PMID: 38149039 PMCID: PMC10751011 DOI: 10.1212/nxg.0000000000200112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/02/2023] [Indexed: 12/28/2023]
Abstract
Background and Objectives A hexanucleotide repeat expansion in the noncoding region of the C9orf72 gene is the most common genetically identifiable cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia in populations of European ancestry. Pedigrees associated with this expansion exhibit phenotypic heterogeneity and incomplete disease penetrance, the basis of which is poorly understood. Relatives of those carrying the C9orf72 repeat expansion exhibit a characteristic cognitive endophenotype independent of carrier status. To examine whether additional shared genetic or environmental risks within kindreds could compel this observation, we have conducted a detailed cross-sectional study of the inheritance within multigenerational Irish kindreds carrying the C9orf72 repeat expansion. Methods One hundred thirty-one familial ALS pedigrees, 59 of which carried the C9orf72 repeat expansion (45.0% [95% CI 36.7-53.5]), were identified through the Irish population-based ALS register. C9orf72 genotyping was performed using repeat-primed PCR with amplicon fragment length analysis. Pedigrees were further investigated using SNP, targeted sequencing data, whole-exome sequencing, and whole-genome sequencing. Results We identified 21 kindreds where at least 1 family member with ALS carried the C9orf72 repeat expansion and from whom DNA was available from multiple affected family members. Of these, 6 kindreds (28.6% [95% CI 11.8-48.3]) exhibited discordant segregation. The C9orf72 haplotype was studied in 2 families and was found to segregate with the C9orf72-positive affected relative but not the C9orf72-negative affected relative. No other ALS pathogenic variants were identified within these discordant kindreds. Discussion Family members of kindreds associated with the C9orf72 repeat expansion may carry an increased risk of developing ALS independent of their observed carrier status. This has implications for assessment and counseling of asymptomatic individuals regarding their genetic risk.
Collapse
Affiliation(s)
- Marie Ryan
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Mark A Doherty
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Ahmad Al Khleifat
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Emmet Costello
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Jennifer C Hengeveld
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Mark Heverin
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Ammar Al-Chalabi
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Russell L Mclaughlin
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| | - Orla Hardiman
- From the Academic Unit of Neurology (M.R., E.C., M.H., O.H.) and Smurfit Institute of Genetics (M.A.D., J.C.H., R.L.M.), Trinity College Dublin, Ireland; Department of Basic and Clinical Neuroscience (A.A., A.A.-C.), Maurice Wohl Clinical Neuroscience Institute, King's College London, United Kingdom; Department of Psychology (E.C.), Beaumont Hospital, Dublin, Ireland; King's College Hospital (A.A.-C.), London, United Kingdom; and Department of Neurology (O.H.), Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
41
|
Limone F, Couto A, Wang JY, Zhang Y, McCourt B, Huang C, Minkin A, Jani M, McNeer S, Keaney J, Gillet G, Gonzalez RL, Goodman WA, Kadiu I, Eggan K, Burberry A. Myeloid and lymphoid expression of C9orf72 regulates IL-17A signaling in mice. Sci Transl Med 2024; 16:eadg7895. [PMID: 38295187 PMCID: PMC11247723 DOI: 10.1126/scitranslmed.adg7895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
A mutation in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Patients with ALS or FTD often develop autoimmunity and inflammation that precedes or coincides with the onset of neurological symptoms, but the underlying mechanisms are poorly understood. Here, we knocked out murine C9orf72 in seven hematopoietic progenitor compartments by conditional mutagenesis and found that myeloid lineage C9orf72 prevents splenomegaly, loss of tolerance, and premature mortality. Furthermore, we demonstrated that C9orf72 plays a role in lymphoid cells to prevent interleukin-17A (IL-17A) production and neutrophilia. Mass cytometry identified early and sustained elevation of the costimulatory molecule CD80 expressed on C9orf72-deficient mouse macrophages, monocytes, and microglia. Enrichment of CD80 was similarly observed in human spinal cord microglia from patients with C9ORF72-mediated ALS compared with non-ALS controls. Single-cell RNA sequencing of murine spinal cord, brain cortex, and spleen demonstrated coordinated induction of gene modules related to antigen processing and presentation and antiviral immunity in C9orf72-deficient endothelial cells, microglia, and macrophages. Mechanistically, C9ORF72 repressed the trafficking of CD80 to the cell surface in response to Toll-like receptor agonists, interferon-γ, and IL-17A. Deletion of Il17a in C9orf72-deficient mice prevented CD80 enrichment in the spinal cord, reduced neutrophilia, and reduced gut T helper type 17 cells. Last, systemic delivery of an IL-17A neutralizing antibody augmented motor performance and suppressed neuroinflammation in C9orf72-deficient mice. Altogether, we show that C9orf72 orchestrates myeloid costimulatory potency and provide support for IL-17A as a therapeutic target for neuroinflammation associated with ALS or FTD.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Leiden University Medical Center, LUMC, 2333 ZA Leiden, The Netherlands
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Jin-Yuan Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Yingying Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Blake McCourt
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Cerianne Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Adina Minkin
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marghi Jani
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sarah McNeer
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - James Keaney
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Gaëlle Gillet
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Rodrigo Lopez Gonzalez
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Irena Kadiu
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Aaron Burberry
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
42
|
Dey B, Kumar A, Patel AB. Pathomechanistic Networks of Motor System Injury in Amyotrophic Lateral Sclerosis. Curr Neuropharmacol 2024; 22:1778-1806. [PMID: 37622689 PMCID: PMC11284732 DOI: 10.2174/1570159x21666230824091601] [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: 04/21/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 08/26/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is the most common, adult-onset, progressive motor neurodegenerative disorder that results in death within 3 years of the clinical diagnosis. Due to the clinicopathological heterogeneity, any reliable biomarkers for diagnosis or prognosis of ALS have not been identified till date. Moreover, the only three clinically approved treatments are not uniformly effective in slowing the disease progression. Over the last 15 years, there has been a rapid advancement in research on the complex pathomechanistic landscape of ALS that has opened up new avenues for successful clinical translation of targeted therapeutics. Multiple studies suggest that the age-dependent interaction of risk-associated genes with environmental factors and endogenous modifiers is critical to the multi-step process of ALS pathogenesis. In this review, we provide an updated discussion on the dysregulated cross-talk between intracellular homeostasis processes, the unique molecular networks across selectively vulnerable cell types, and the multisystemic nature of ALS pathomechanisms. Importantly, this work highlights the alteration in epigenetic and epitranscriptomic landscape due to gene-environment interactions, which have been largely overlooked in the context of ALS pathology. Finally, we suggest that precision medicine research in ALS will be largely benefitted from the stratification of patient groups based on the clinical phenotype, onset and progression, genome, exposome, and metabolic identities.
Collapse
Affiliation(s)
- Bedaballi Dey
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Arvind Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Anant Bahadur Patel
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
43
|
Baumgartner D, Mušová Z, Zídková J, Hedvičáková P, Vlčková E, Joppeková L, Kramářová T, Fajkusová L, Stránecký V, Geryk J, Votýpka P, Mazanec R. Genetic Landscape of Amyotrophic Lateral Sclerosis in Czech Patients. J Neuromuscul Dis 2024; 11:1035-1048. [PMID: 39058450 PMCID: PMC11380243 DOI: 10.3233/jnd-230236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Background Genetic factors are involved in the pathogenesis of familial and sporadic amyotrophic lateral sclerosis (ALS) and constitute a link to its association with frontotemporal dementia (FTD). Gene-targeted therapies for some forms of ALS (C9orf72, SOD1) have recently gained momentum. Genetic architecture in Czech ALS patients has not been comprehensively assessed so far. Objective We aimed to deliver pilot data on the genetic landscape of ALS in our country. Methods A cohort of patients with ALS (n = 88), recruited from two Czech Neuromuscular Centers, was assessed for hexanucleotide repeat expansion (HRE) in C9orf72 and also for genetic variations in other 36 ALS-linked genes via next-generation sequencing (NGS). Nine patients (10.1%) had a familial ALS. Further, we analyzed two subgroups of sporadic patients - with concomitant FTD (n = 7) and with young-onset of the disease (n = 22). Results We detected the pathogenic HRE in C9orf72 in 12 patients (13.5%) and three other pathogenic variants in FUS, TARDBP and TBK1, each in one patient. Additional 7 novel and 9 rare known variants with uncertain causal significance have been detected in 15 patients. Three sporadic patients with FTD (42.9%) were harbouring a pathogenic variant (all HRE in C9orf72). Surprisingly, none of the young-onset sporadic patients harboured a pathogenic variant and we detected no pathogenic SOD1 variant in our cohort. Conclusion Our findings resemble those from other European populations, with the highest prevalence of HRE in the C9orf72 gene. Further, our findings suggest a possibility of a missing genetic variability among young-onset patients.
Collapse
Affiliation(s)
- Daniel Baumgartner
- Department of Neurology, Neuromuscular Center, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Zuzana Mušová
- Department of Biology and Medical Genetics, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Jana Zídková
- Center for Molecular Biology and Genetics, Internal Haematology and Oncology Clinic, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Petra Hedvičáková
- Department of Biology and Medical Genetics, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Eva Vlčková
- Department of Neurology, Neuromuscular Center, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lubica Joppeková
- Department of Neurology, Neuromuscular Center, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tereza Kramářová
- Center for Molecular Biology and Genetics, Internal Haematology and Oncology Clinic, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Lenka Fajkusová
- Center for Molecular Biology and Genetics, Internal Haematology and Oncology Clinic, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Viktor Stránecký
- Department of Pediatrics and Inherited Metabolic Disorders, Research Unit for Rare Diseases, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Jan Geryk
- Department of Biology and Medical Genetics, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Pavel Votýpka
- Department of Biology and Medical Genetics, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Radim Mazanec
- Department of Neurology, Neuromuscular Center, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
44
|
Martinelli I, Ghezzi A, Zucchi E, Gianferrari G, Ferri L, Moglia C, Manera U, Solero L, Vasta R, Canosa A, Grassano M, Brunetti M, Mazzini L, De Marchi F, Simonini C, Fini N, Vinceti M, Pinti M, Chiò A, Calvo A, Mandrioli J. Predictors for progression in amyotrophic lateral sclerosis associated to SOD1 mutation: insight from two population-based registries. J Neurol 2023; 270:6081-6092. [PMID: 37668704 DOI: 10.1007/s00415-023-11963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Uncovering distinct features and trajectories of amyotrophic lateral sclerosis (ALS) associated with SOD1 mutations (SOD1-ALS) can provide valuable insights for patient' counseling and stratification for trials, and interventions timing. Our study aims to pinpoint distinct clinical characteristics of SOD1-ALS by delving into genotype-phenotype correlations and factors that potentially impact disease progression. METHODS This is a retrospective observational study of a SOD1-ALS cohort from two Italian registers situated in the regions of Emilia-Romagna, Piedmont and Valle d'Aosta. RESULTS Out of 2204 genotyped ALS patients, 2.5% carried SOD1 mutations, with a M:F ratio of 0.83. SOD1-ALS patients were younger, and more frequently reported a family history of ALS and/or FTD. SOD1-ALS had a longer survival compared to patients without ALS-associated gene mutations. However, here was considerable variability in survival across distinct SOD1 mutations, with an average survival of less than a year for the L39V, G42S, G73S, D91N mutations. Among SOD1-ALS, multivariate analysis showed that, alongside established clinical prognostic factors such as advanced age at onset and high progression rate at diagnosis, mutations located in exon 2 or within highly conserved gene positions predicted worse survival. Conversely, among comorbidities, cancer history was independently associated with longer survival. INTERPRETATION Within the context of an overall slower disease, SOD1-ALS exhibits some degree of heterogeneity linked to the considerable genetic diversity arising from the multitude of potential mutations sites and specific clinical prognostic factors, including cancer history. Revealing the factors that modulate the phenotypic heterogeneity of SOD1-ALS could prove advantageous in improving the efficacy of upcoming therapeutic approaches.
Collapse
Affiliation(s)
- Ilaria Martinelli
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Clinical and Experimental Medicine Ph.D. Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Ghezzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Zucchi
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy.
- Neuroscience PhD Program, University of Modena and Reggio Emilia, Modena, Italy.
| | - Giulia Gianferrari
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Ferri
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Neuroscience PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Moglia
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Umberto Manera
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Luca Solero
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Rosario Vasta
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Antonio Canosa
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Maurizio Grassano
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Maura Brunetti
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Letizia Mazzini
- Neurology Unit, ALS Center, AOU Maggiore della Carità and University of Piemonte Orientale, Novara, Italy
| | - Fabiola De Marchi
- Neurology Unit, ALS Center, AOU Maggiore della Carità and University of Piemonte Orientale, Novara, Italy
| | - Cecilia Simonini
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
| | - Nicola Fini
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
| | - Marco Vinceti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Science of Public Health, Research Centre in Environmental, Genetic and Nutritional Epidemiology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Adriano Chiò
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Andrea Calvo
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Jessica Mandrioli
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
45
|
Mitra S, Dash R, Nishan AA, Habiba SU, Moon IS. Brain modulation by the gut microbiota: From disease to therapy. J Adv Res 2023; 53:153-173. [PMID: 36496175 PMCID: PMC10658262 DOI: 10.1016/j.jare.2022.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The gut microbiota (GM) and brain are strongly associated, which significantly affects neuronal development and disorders. GM-derived metabolites modulate neuronal function and influence many cascades in age-related neurodegenerative disorders (NDDs). Because of the dual role of GM in neuroprotection and neurodegeneration, understanding the balance between beneficial and harmful bacteria is crucial for applying this approach to clinical therapies. AIM OF THE REVIEW This review briefly discusses the role of the gut-brain relationship in promoting brain and cognitive function. Although a healthy gut environment is helpful for brain function, gut dysbiosis can disrupt the brain's environment and create a vicious cycle of degenerative cascades. The ways in which the GM population can affect brain function and the development of neurodegeneration are also discussed. In the treatment and management of NDDs, the beneficial effects of methods targeting GM populations and their derivatives, including probiotics, prebiotics, and fecal microbial transplantation (FMT) are also highlighted. KEY SCIENTIFIC CONCEPT OF THE REVIEW In this review, we aimed to provide a deeper understanding of the mechanisms of the gut microbe-brain relationship and their twin roles in neurodegeneration progression and therapeutic applications. Here, we attempted to highlight the different pathways connecting the brain and gut, together with the role of GM in neuroprotection and neuronal development. Furthermore, potential roles of GM metabolites in the pathogenesis of brain disorders and in strategies for its treatment are also investigated. By analyzing existing in vitro, in vivo and clinical studies, this review attempts to identify new and promising therapeutic strategies for central nervous system (CNS) disorders. As the connection between the gut microbe-brain relationship and responses to NDD treatments is less studied, this review will provide new insights into the global mechanisms of GM modulation in disease progression, and identify potential future perspectives for developing new therapies to treat NDDs.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Amena Al Nishan
- Department of Medicine, Chittagong Medical College, Chittagong 4203, Bangladesh
| | - Sarmin Ummey Habiba
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea.
| |
Collapse
|
46
|
Roggenbuck J, Eubank BHF, Wright J, Harms MB, Kolb SJ. Evidence-based consensus guidelines for ALS genetic testing and counseling. Ann Clin Transl Neurol 2023; 10:2074-2091. [PMID: 37691292 PMCID: PMC10646996 DOI: 10.1002/acn3.51895] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/12/2023] [Indexed: 09/12/2023] Open
Abstract
OBJECTIVE Advances in amyotrophic lateral sclerosis (ALS) gene discovery, ongoing gene therapy trials, and patient demand have driven increased use of ALS genetic testing. Despite this progress, the offer of genetic testing to persons with ALS is not yet "standard of care." Our primary goal is to develop clinical ALS genetic counseling and testing guidelines to improve and standardize genetic counseling and testing practice among neurologists, genetic counselors or any provider caring for persons with ALS. METHODS Core clinical questions were identified and a rapid review performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA-P) 2015 method. Guideline recommendations were drafted and the strength of evidence for each recommendation was assessed by combining two systems: the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) System and the Evaluation of Genomic Applications in Practice and Prevention (EGAPP). A modified Delphi approach was used to reach consensus among a group of content experts for each guideline statement. RESULTS A total of 35 guideline statements were developed. In summary, all persons with ALS should be offered single-step genetic testing, consisting of a C9orf72 assay, along with sequencing of SOD1, FUS, and TARDBP, at a minimum. The key education and genetic risk assessments that should be provided before and after testing are delineated. Specific guidance regarding testing methods and reporting for C9orf72 and other genes is provided for commercial laboratories. INTERPRETATION These evidence-based, consensus guidelines will support all stakeholders in the ALS community in navigating benefits and challenges of genetic testing.
Collapse
Affiliation(s)
- Jennifer Roggenbuck
- Division of Human Genetics, Department of Internal MedicineThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Breda H. F. Eubank
- Health & Physical Education Department, Faculty of Health, Community, & EducationMount Royal University4825 Mount Royal Gate SWCalgaryAlbertaCanada
| | - Joshua Wright
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Matthew B. Harms
- Department of NeurologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
| | - Stephen J. Kolb
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Department of Biological Chemistry & PharmacologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
47
|
Oprisan AL, Popescu BO. Dysautonomia in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:14927. [PMID: 37834374 PMCID: PMC10573406 DOI: 10.3390/ijms241914927] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease, characterized in its typical presentation by a combination of lower and upper motor neuron symptoms, with a progressive course and fatal outcome. Due to increased recognition of the non-motor symptoms, it is currently considered a multisystem disorder with great heterogeneity, regarding genetical, clinical, and neuropathological features. Often underestimated, autonomic signs and symptoms have been described in patients with ALS, and various method analyses have been used to assess autonomic nervous system involvement. The aim of this paper is to offer a narrative literature review on autonomic disturbances in ALS, based on the scarce data available to date.
Collapse
Affiliation(s)
- Alexandra L Oprisan
- Department of Clinical Neurosciences, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Neurology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
48
|
Goutman SA, Savelieff MG, Jang DG, Hur J, Feldman EL. The amyotrophic lateral sclerosis exposome: recent advances and future directions. Nat Rev Neurol 2023; 19:617-634. [PMID: 37709948 PMCID: PMC11027963 DOI: 10.1038/s41582-023-00867-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease of motor neuron degeneration with typical survival of only 2-5 years from diagnosis. The causes of ALS are multifactorial: known genetic mutations account for only around 70% of cases of familial ALS and 15% of sporadic cases, and heritability estimates range from 8% to 61%, indicating additional causes beyond genetics. Consequently, interest has grown in environmental contributions to ALS risk and progression. The gene-time-environment hypothesis posits that ALS onset occurs through an interaction of genes with environmental exposures during ageing. An alternative hypothesis, the multistep model of ALS, suggests that several hits, at least some of which could be environmental, are required to trigger disease onset, even in the presence of highly penetrant ALS-associated mutations. Studies have sought to characterize the ALS exposome - the lifetime accumulation of environmental exposures that increase disease risk and affect progression. Identifying the full scope of environmental toxicants that enhance ALS risk raises the prospect of preventing disease by eliminating or mitigating exposures. In this Review, we summarize the evidence for an ALS exposome, discussing the strengths and limitations of epidemiological studies that have identified contributions from various sources. We also consider potential mechanisms of exposure-mediated toxicity and suggest future directions for ALS exposome research.
Collapse
Affiliation(s)
- Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Masha G Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Dae-Gyu Jang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Solovyev N, Lucio M, Mandrioli J, Forcisi S, Kanawati B, Uhl J, Vinceti M, Schmitt-Kopplin P, Michalke B. Interplay of Metallome and Metabolome in Amyotrophic Lateral Sclerosis: A Study on Cerebrospinal Fluid of Patients Carrying Disease-Related Gene Mutations. ACS Chem Neurosci 2023; 14:3035-3046. [PMID: 37608584 PMCID: PMC10485893 DOI: 10.1021/acschemneuro.3c00128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a lethal progressive neurodegenerative disease, characterized by a loss of function of upper and lower motor neurons. This study aimed to explore probable pathological alterations occurring in individuals with ALS compared to neurologically healthy controls through the analysis of cerebrospinal fluid (CSF), a medium, which directly interacts with brain parenchyma. A total of 7 ALS patients with disease-associated mutations (ATXN2, C9ORF72, FUS, SOD1, and TARDBP) and 13 controls were included in the study. Multiple analytical approaches were employed, including metabolomic and metallomics profiling, as well as genetic screening, using CSF samples obtained from the brain compartment. Data analysis involved the application of multivariate statistical methods. Advanced hyphenated selenium and redox metal (iron, copper, and manganese) speciation techniques and nontargeted Fourier transform ion cyclotron resonance mass spectrometry-based metabolomics were used for data acquisition. Nontargeted metabolomics showed reduced steroids, including sex hormones; additionally, copper and manganese species were found to be the most relevant features for ALS patients. This indicates a potential alteration of sex hormone pathways in the ALS-affected brain, as reflected in the CSF.
Collapse
Affiliation(s)
- Nikolay Solovyev
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Marianna Lucio
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Jessica Mandrioli
- Department
of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department
of Neurosciences, Azienda Ospedaliero Universitaria
di Modena, 41126 Modena, Italy
| | - Sara Forcisi
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Basem Kanawati
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Jenny Uhl
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Marco Vinceti
- CREAGEN
Research Center of Environmental, Genetic and Nutritional Epidemiology,
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Philippe Schmitt-Kopplin
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Bernhard Michalke
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| |
Collapse
|
50
|
Davidson JM, Wu SSL, Rayner SL, Cheng F, Duncan K, Russo C, Newbery M, Ding K, Scherer NM, Balez R, García-Redondo A, Rábano A, Rosa-Fernandes L, Ooi L, Williams KL, Morsch M, Blair IP, Di Ieva A, Yang S, Chung RS, Lee A. The E3 Ubiquitin Ligase SCF Cyclin F Promotes Sequestosome-1/p62 Insolubility and Foci Formation and is Dysregulated in ALS and FTD Pathogenesis. Mol Neurobiol 2023; 60:5034-5054. [PMID: 37243816 PMCID: PMC10415446 DOI: 10.1007/s12035-023-03355-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 04/15/2023] [Indexed: 05/29/2023]
Abstract
Amyotrophic lateral sclerosis (ALS)- and frontotemporal dementia (FTD)-linked mutations in CCNF have been shown to cause dysregulation to protein homeostasis. CCNF encodes for cyclin F, which is part of the cyclin F-E3 ligase complex SCFcyclinF known to ubiquitylate substrates for proteasomal degradation. In this study, we identified a function of cyclin F to regulate substrate solubility and show how cyclin F mechanistically underlies ALS and FTD disease pathogenesis. We demonstrated that ALS and FTD-associated protein sequestosome-1/p62 (p62) was a canonical substrate of cyclin F which was ubiquitylated by the SCFcyclinF complex. We found that SCFcyclin F ubiquitylated p62 at lysine(K)281, and that K281 regulated the propensity of p62 to aggregate. Further, cyclin F expression promoted the aggregation of p62 into the insoluble fraction, which corresponded to an increased number of p62 foci. Notably, ALS and FTD-linked mutant cyclin F p.S621G aberrantly ubiquitylated p62, dysregulated p62 solubility in neuronal-like cells, patient-derived fibroblasts and induced pluripotent stem cells and dysregulated p62 foci formation. Consistently, motor neurons from patient spinal cord tissue exhibited increased p62 ubiquitylation. We suggest that the p.S621G mutation impairs the functions of cyclin F to promote p62 foci formation and shift p62 into the insoluble fraction, which may be associated to aberrant mutant cyclin F-mediated ubiquitylation of p62. Given that p62 dysregulation is common across the ALS and FTD spectrum, our study provides insights into p62 regulation and demonstrates that ALS and FTD-linked cyclin F mutant p.S621G can drive p62 pathogenesis associated with ALS and FTD.
Collapse
Affiliation(s)
- Jennilee M Davidson
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia.
| | - Sharlynn S L Wu
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Stephanie L Rayner
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Flora Cheng
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Kimberley Duncan
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Carlo Russo
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Michelle Newbery
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Kunjie Ding
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Natalie M Scherer
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Rachelle Balez
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Alberto García-Redondo
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER U-723), Unidad de ELA, Instituto de Investigación Hospital 12 de Octubre de Madrid, SERMAS, Madrid, Spain
| | - Alberto Rábano
- Neuropathology Department and CIEN Tissue Bank, Alzheimer's Centre Reina Sofia-CIEN Foundation, 28031, Madrid, Spain
| | - Livia Rosa-Fernandes
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Kelly L Williams
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Marco Morsch
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Ian P Blair
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Antonio Di Ieva
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Shu Yang
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Roger S Chung
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Albert Lee
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Road, Sydney, NSW, 2109, Australia
| |
Collapse
|