1
|
Capelle DP, Sabirin W, Zulhairy-Liong NA, Edgar S, Goh KJ, Ahmad-Annuar A, Shahrizaila N. Multistep modeling applied to a Malaysian ALS registry. Amyotroph Lateral Scler Frontotemporal Degener 2025; 26:157-161. [PMID: 39363643 DOI: 10.1080/21678421.2024.2410280] [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: 07/12/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
OBJECTIVE To apply the multistep model of pathogenesis in amyotrophic lateral sclerosis (ALS) to data from a multiethnic Malaysian registry. METHODS Clinical data, including age at symptom onset, was collected from 289 patients who presented to our multidisciplinary clinic from 2016 until 2024. A least squares linear regression model was constructed from the logarithm of approximated incidence and the logarithm of age. Population incidence was approximated by adjusting the absolute numbers of patients in 5 year groups by the size of the general population in the respective age group. RESULTS A linear relationship between log of incidence versus log of age was observed, with a slope of 4.57 (95% CI, 3.3-5.8) and an r2 value of 0.93, suggesting a 6-step process. CONCLUSION Progression toward symptom onset in Malaysian ALS patients appears consistent with a multistep model of disease as observed in other cohorts.
Collapse
Affiliation(s)
- David Paul Capelle
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia and
| | - Wafa Sabirin
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia and
| | | | - Suzanna Edgar
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia and
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Khean-Jin Goh
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia and
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nortina Shahrizaila
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia and
| |
Collapse
|
2
|
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
|
3
|
Ye L, Dittlau KS, Sicart A, Janky R, Van Damme P, Van Den Bosch L. Sporadic ALS iPSC-derived motor neurons show axonal defects linked to altered axon guidance pathways. Neurobiol Dis 2025:106815. [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] [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
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Santurtún A, Medín P, Riancho JA, Santiago-Setién M, Ortiz F, López de Munain A, Almendra R, Riancho J. Temporo-spatial analysis of amyotrophic lateral sclerosis in Spain: Altitude and land use as new determinants of the disease. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177796. [PMID: 39616922 DOI: 10.1016/j.scitotenv.2024.177796] [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: 10/16/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is the most common neurodegenerative disease affecting motor neurons. Currently, ALS is conceived as the result of the interaction between genetics, environmental factors, and aging. This study analyzed the spatial and temporal patterns of ALS in Spain, delving into the potential relationships between altitude, land cover, and this disease. METHODOLOGY ALS death data were collected over a 19-year period, including information on sex, age and municipality of residence. The standardized mortality rate was calculated for each municipality of residencia, and Anselin's local Moran's I statistic was used to identify clusters of high and low incidence. Altitude data were sourced from the Copernicus Land Monitoring Services, while land cover data came from CORINE satellite images and national agricultural statistics. RESULTS The average annual incidence of ALS deaths among adults was 2.5 per 100,000 people. Higher mortality rates were noted in males (2.8) than in females (2.3), with both sexes exhibiting a rising mortality trend in a temporal analysis. Cluster analysis revealed that high mortality areas were mostly located in the North and Northeast of the country. Municipalities in these clusters had significantly lower median altitudes and larger areas of Permanently Irrigated Arable Land and Broad-Leaved Forest. CONCLUSION This study provides new evidence about the increase in ALS cases in European countries during the last decades, reporting for the first time altitude and certain agricultural land uses as potential geographic determinants of the disease.
Collapse
Affiliation(s)
- Ana Santurtún
- Unit of Legal Medicine, Department of Physiology and Pharmacology, University of Cantabria, Santander, Spain; Valdecilla Research Institute (IDIVAL), Santander, Spain.
| | - Pablo Medín
- Unit of Legal Medicine, Department of Physiology and Pharmacology, University of Cantabria, Santander, Spain
| | - José Antonio Riancho
- Valdecilla Research Institute (IDIVAL), Santander, Spain; Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain; Hospital U. M. Valdecilla, University of Cantabria, Santander, Spain
| | | | - Fernando Ortiz
- Hospital U. M. Valdecilla, University of Cantabria, Santander, Spain
| | - Adolfo López de Munain
- Neurology Department, Donostia University Hospital, Spain; University of the Basque Country-Osakidetza, Spain; CIBERNED, Barcelona, Spain; Biodonostia, 20014 Donostia-San Sebastián, Spain
| | - Ricardo Almendra
- Centre of Studies on Geography and Spatial Planning (CEGOT), Coimbra, Portugal; Department of Geography and Tourism, Faculty of Arts and Humanities, Colégio de São Jerónimo, University of Coimbra, Coimbra, Portugal
| | - Javier Riancho
- Valdecilla Research Institute (IDIVAL), Santander, Spain; Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain; CIBERNED, Barcelona, Spain; Hospital Sierrallana, Torrelavega, Spain.
| |
Collapse
|
8
|
Frolov A, D'sa E, Henderson C, Guzman MA, Hayat G, Martin JR. Complex Genetic Framework in Familial Amyotrophic Lateral Sclerosis With a C9ORF72 Mutation: A Case Report. Cureus 2024; 16:e76027. [PMID: 39835009 PMCID: PMC11743604 DOI: 10.7759/cureus.76027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
A significantly diverse clinical presentation of amyotrophic lateral sclerosis (ALS), even in its best-studied familial form, continues to hinder current efforts to develop effective disease-modifying drugs for the cure of this rapidly progressive, fatal neuromuscular disease. We have previously shown that clinical heterogeneity of sporadic ALS (sALS) could be explained, at least in part, by its polygenic nature as well as by the presence of mutated genes linked to non-ALS neurological diseases and genes known to mediate ALS-related pathologies. We hypothesized that a similar genetic framework could also be present in patients with familial ALS (fALS). To test this hypothesis, we conducted post-mortem genetic screening of an individual with fALS and a mutation in the C9ORF72 gene. C9ORF72 mutations are highly penetrant and are present in the majority of fALS patients. Genetic screening by whole exome sequencing (WES) on the next generation sequencing (NGS) Illumina platform (San Diego, CA, USA) followed by examination of the respective rare (minor allele frequency (MAF) ≤ 0.01) pathological/deleterious genetic variants yielded results consistent with our hypothesis of the presence of a complex genetic framework in fALS. Additional members of this genetic framework were identified when the low-frequency (0.01 < MAF < 0.05) pathological/deleterious genetic variants were analyzed with the low-frequency biallelic AHNAK2, GLI3, PTIRM1, and ZNF254 variants, warranting a closer look at their potentially important role in fALS as C9ORF72 genetic modifiers as well as their link to both neuromuscular disorders/ALS and cancer. Therefore, in addition to the current genetic screening using a standard panel of ALS-related genes, a supplementary screening by WES could be very beneficial for the development of personalized treatment of ALS patients as well as in search of the respective efficient disease-modifying drugs.
Collapse
Affiliation(s)
- Andrey Frolov
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, St. Louis, USA
| | - Elizabeth D'sa
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, St. Louis, USA
| | - Camille Henderson
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, St. Louis, USA
| | - Miguel A Guzman
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, USA
| | - Ghazala Hayat
- Department of Neurology, Saint Louis University School of Medicine, St. Louis, USA
| | - John R Martin
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, St. Louis, USA
| |
Collapse
|
9
|
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
|
10
|
Saez-Atienzar S, Souza CDS, Chia R, Beal SN, Lorenzini I, Huang R, Levy J, Burciu C, Ding J, Gibbs JR, Jones A, Dewan R, Pensato V, Peverelli S, Corrado L, van Vugt JJFA, van Rheenen W, Tunca C, Bayraktar E, Xia M, Iacoangeli A, Shatunov A, Tiloca C, Ticozzi N, Verde F, Mazzini L, Kenna K, Al Khleifat A, Opie-Martin S, Raggi F, Filosto M, Piccinelli SC, Padovani A, Gagliardi S, Inghilleri M, Ferlini A, Vasta R, Calvo A, Moglia C, Canosa A, Manera U, Grassano M, Mandrioli J, Mora G, Lunetta C, Tanel R, Trojsi F, Cardinali P, Gallone S, Brunetti M, Galimberti D, Serpente M, Fenoglio C, Scarpini E, Comi GP, Corti S, Del Bo R, Ceroni M, Pinter GL, Taroni F, Bella ED, Bersano E, Curtis CJ, Lee SH, Chung R, Patel H, Morrison KE, Cooper-Knock J, Shaw PJ, Breen G, Dobson RJB, Dalgard CL, Scholz SW, Al-Chalabi A, van den Berg LH, McLaughlin R, Hardiman O, Cereda C, Sorarù G, D'Alfonso S, Chandran S, Pal S, Ratti A, Gellera C, Johnson K, Doucet-O'Hare T, Pasternack N, Wang T, Nath A, Siciliano G, Silani V, Başak AN, Veldink JH, Camu W, Glass JD, Landers JE, Chiò A, Sattler R, Shaw CE, Ferraiuolo L, Fogh I, Traynor BJ. Mechanism-free repurposing of drugs for C9orf72-related ALS/FTD using large-scale genomic data. CELL GENOMICS 2024; 4:100679. [PMID: 39437787 PMCID: PMC11605688 DOI: 10.1016/j.xgen.2024.100679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 09/22/2024] [Indexed: 10/25/2024]
Abstract
Repeat expansions in the C9orf72 gene are the most common genetic cause of (ALS) and frontotemporal dementia (FTD). Like other genetic forms of neurodegeneration, pinpointing the precise mechanism(s) by which this mutation leads to neuronal death remains elusive, and this lack of knowledge hampers the development of therapy for C9orf72-related disease. We used an agnostic approach based on genomic data (n = 41,273 ALS and healthy samples, and n = 1,516 C9orf72 carriers) to overcome these bottlenecks. Our drug-repurposing screen, based on gene- and expression-pattern matching and information about the genetic variants influencing onset age among C9orf72 carriers, identified acamprosate, a γ-aminobutyric acid analog, as a potentially repurposable treatment for patients carrying C9orf72 repeat expansions. We validated its neuroprotective effect in cell models and showed comparable efficacy to riluzole, the current standard of care. Our work highlights the potential value of genomics in repurposing drugs in situations where the underlying pathomechanisms are inherently complex. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Sara Saez-Atienzar
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Ohio State University, Columbus, OH 43210, USA.
| | - Cleide Dos Santos Souza
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ruth Chia
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Selina N Beal
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ileana Lorenzini
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Jennifer Levy
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Camelia Burciu
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - J Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Ashley Jones
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ramita Dewan
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Viviana Pensato
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Peverelli
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Lucia Corrado
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Joke J F A van Vugt
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ceren Tunca
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Elif Bayraktar
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Cinzia Tiloca
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Letizia Mazzini
- Amyotrophic Lateral Sclerosis Center, Department of Neurology "Maggiore della Carità" University Hospital, Novara, Italy
| | - Kevin Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Sarah Opie-Martin
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Flavia Raggi
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Massimiliano Filosto
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Cotti Piccinelli
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stella Gagliardi
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Maurizio Inghilleri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, 00185 Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Science, University of Ferrara, Ferrara, Italy
| | - Rosario Vasta
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Andrea Calvo
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Cristina Moglia
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Antonio Canosa
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Umberto Manera
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Maurizio Grassano
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Gabriele Mora
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Christian Lunetta
- Department of Neurorehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Institute of Milan, Milan, Italy; NEMO Clinical Center Milano, Fondazione Serena Onlus, Milan, Italy
| | - Raffaella Tanel
- Operative Unit of Neurology, S. Chiara Hospital, Trento, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | | | - Salvatore Gallone
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Maura Brunetti
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Maria Serpente
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Elio Scarpini
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo P Comi
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberto Del Bo
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Ceroni
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Giuseppe Lauria Pinter
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eleonora Dalla Bella
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
| | - Enrica Bersano
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; "L. Sacco" Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Charles J Curtis
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Sang Hyuck Lee
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Raymond Chung
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Hamel Patel
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Karen E Morrison
- School of Medicine, Dentistry, and Biomedical Sciences, Faculty of Medicine Health and Life Sciences, Queen's University, Belfast, UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Gerome Breen
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Richard J B Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK; NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Health Data Research UK London, University College London, London, UK; Institute of Health Informatics, University College London, London, UK; NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, London, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, King's College Hospital, London SE5 9RS, UK
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Russell McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Siddharthan Chandran
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Suvankar Pal
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; Centre for Neuroregeneration and Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Kory Johnson
- Bioinformatics Section, Information Technology Program (ITP), Division of Intramural Research (DIR), National Institute of Neurological Disorders & Stroke, NIH, Bethesda, MD 20892, USA
| | - Tara Doucet-O'Hare
- Neuro-oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Nicholas Pasternack
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Tongguang Wang
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Avindra Nath
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Ayşe Nazlı Başak
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - William Camu
- ALS Center, CHU Gui de Chauliac, University of Montpellier, Montpellier, France
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Rita Sattler
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christopher E Shaw
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Isabella Fogh
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK; National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; RNA Therapeutics Laboratory, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA.
| |
Collapse
|
11
|
Theme 5 Human Cell Biology and Pathology. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:158-184. [PMID: 39508672 DOI: 10.1080/21678421.2024.2403302] [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]
|
12
|
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]
|
13
|
Sun S, Chen Y, Yun Y, Zhao B, Ren Q, Sun X, Meng X, Yan C, Lin P, Liu S. Elevated peripheral inflammation is associated with choroid plexus enlargement in independent sporadic amyotrophic lateral sclerosis cohorts. Fluids Barriers CNS 2024; 21:83. [PMID: 39434103 PMCID: PMC11492712 DOI: 10.1186/s12987-024-00586-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: 07/31/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Using neuroimaging techniques, growing evidence has suggested that the choroid plexus (CP) volume is enlarged in multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Notably, the CP has been suggested to play an important role in inflammation-induced CNS damage under disease conditions. However, to our knowledge, no study has investigated the relationships between peripheral inflammation and CP volume in sporadic ALS patients. Thus, in this study, we aimed to verify CP enlargement and explore its association with peripheral inflammation in vivo in independent ALS cohorts. METHODS Based on structural MRI data, CP volume was measured using Gaussian mixture models and further manually corrected in two independent cohorts of sporadic ALS patients and healthy controls (HCs). Serum inflammatory protein levels were measured using a novel high-sensitivity Olink proximity extension assay (PEA) technique. Xtreme gradient boosting (XGBoost) was used to explore the contribution of peripheral inflammatory factors to CP enlargement. Then, partial correlation analyses were performed. RESULTS CP volumes were significantly higher in ALS patients than in HCs in the independent cohorts. Compared with HCs, serum levels of CRP, IL-6, CXCL10, and 35 other inflammatory factors were significantly increased in ALS patients. Using the XGBoost approach, we established a model-based importance of features, and the top three predictors of CP volume in ALS patients were CRP, IL-6, and CXCL10 (with gains of 0.24, 0.18, and 0.15, respectively). Correlation analyses revealed that CRP, IL-6, and CXCL10 were significantly associated with CP volume in ALS patients (r = 0.462 ∼ 0.636, p < 0.001). CONCLUSION Our study is the first to reveal a consistent and replicable contribution of peripheral inflammation to CP enlargement in vivo in sporadic ALS patients. Given that CP enlargement has been recently detected in other brain diseases, these findings should consider extending to other disease conditions with a peripheral inflammatory component.
Collapse
Affiliation(s)
- Sujuan Sun
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
| | - Yujing Chen
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
| | - Yan Yun
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Bing Zhao
- Department of Neurology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Qingguo Ren
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Xiaohan Sun
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
| | - Xiangshui Meng
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Chuanzhu Yan
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Pengfei Lin
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China.
| | - Shuangwu Liu
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Qilu Hospital of Shandong University, West Wenhua Street No.107, Jinan, 250012, Shandong, China.
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China.
| |
Collapse
|
14
|
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
|
15
|
Calma AD, Pavey N, Menon P, Vucic S. Neuroinflammation in amyotrophic lateral sclerosis: pathogenic insights and therapeutic implications. Curr Opin Neurol 2024; 37:585-592. [PMID: 38775138 DOI: 10.1097/wco.0000000000001279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
PURPOSE OF REVIEW Neuroinflammation appears to be an important pathogenic process in amyotrophic lateral sclerosis (ALS). Dysfunction of central immune pathways, including activation of microglia and astrocytes, and peripherally derived immune cells, initiate noncell autonomous inflammatory mechanisms leading to degeneration. Cell autonomous pathways linked to ALS genetic mutations have been recently identified as contributing mechanism for neurodegeneration. The current review provides insights into the pathogenic importance of central and peripheral inflammatory processes in ALS pathogenesis and appraises their potential as therapeutic targets. RECENT FINDINGS ALS is a multistep process mediated by a complex interaction of genetic, epigenetic, and environmental factors. Noncell autonomous inflammatory pathways contribute to neurodegeneration in ALS. Activation of microglia and astrocytes, along with central nervous system infiltration of peripherally derived pro-inflammatory innate (NK-cells/monocytes) and adaptive (cell-mediated/humoral) immune cells, are characteristic of ALS. Dysfunction of regulatory T-cells, elevation of pro-inflammatory cytokines and dysbiosis of gut microbiome towards a pro-inflammatory phenotype, have been reported as pathogenic mechanisms in ALS. SUMMARY Dysregulation of adaptive and innate immunity is pathogenic in ALS, being associated with greater disease burden, more rapid disease course and reduced survival. Strategies aimed at modulating the pro-inflammatory immune components could be of therapeutic utility.
Collapse
Affiliation(s)
- Aicee D Calma
- Brain and Nerve Research Centre, Concord Clinical School, The University of Sydney, Concord Hospital, Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
16
|
Sun S, Chen Y, Zhao B, Zhu J, Wen T, Peng B, Ren Q, Sun X, Lin P, Zhang D, Liu S. Abnormal brain functional network dynamics in amyotrophic lateral sclerosis patients with depression. Brain Imaging Behav 2024; 18:1034-1043. [PMID: 38814545 DOI: 10.1007/s11682-024-00896-5] [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] [Accepted: 05/11/2024] [Indexed: 05/31/2024]
Abstract
Since depression is common in amyotrophic lateral sclerosis (ALS) patients, we aimed to explore the specific brain functional network dynamics in ALS patients with depression (ALS-D) compared with healthy controls (HCs) and ALS patients without depressive symptoms (ALS-ND). According to the DSM-V, 32 ALS-D patients were selected from a large and newly diagnosed ALS cohort. Then, 32 demographic- and cognitive-matched ALS-ND patients were also selected, and 64 HCs were recruited. These participants underwent resting-state fMRI scans, and functional connectivity state analysis and dynamic graph theory were applied to evaluate brain functional network dynamics. Moreover, the Hamilton Depression Rating Scale (HDRS) was used to quantify depressive symptoms in the ALS-D patients. Four distinct states were identified in the ALS-D patients and controls. Compared with that in HCs, the fraction rate (FR) in state 2 was significantly decreased in ALS-D patients, and the FR in state 4 was significantly increased in ALS-D patients. Compared with that of HCs, the dwell time in state 4 was significantly increased in the ALS-D patients. Moreover, compared with that in the ALS-D patients, the FR in state 3 was significantly decreased in the ALS-ND patients. Among the ALS-D patients, there was the suggestion of a positive association between HDRS scores and dwell time of state 4, but this association did not reach statistical significance (r = 0.354; p = 0.055). Depression is an important feature of ALS patients, and we found a special pattern of brain functional network dynamics in ALS-D patients. Our findings may play an important role in understanding the mechanism underlying depression in ALS patients and help develop therapeutic interventions for depressed ALS patients.
Collapse
Affiliation(s)
- Sujuan Sun
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China
| | - Yujing Chen
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China
| | - Bing Zhao
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Jun Zhu
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China
| | - Tianrui Wen
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China
| | - Bingnan Peng
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China
| | - Qingguo Ren
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xiaohan Sun
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China
| | - Pengfei Lin
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China
| | - Dong Zhang
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China.
| | - Shuangwu Liu
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, West Wenhua Street No. 107, Jinan, 250012, China.
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China.
| |
Collapse
|
17
|
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
|
18
|
van der Geest AT, Jakobs CE, Ljubikj T, Huffels CFM, Cañizares Luna M, Vieira de Sá R, Adolfs Y, de Wit M, Rutten DH, Kaal M, Zwartkruis MM, Carcolé M, Groen EJN, Hol EM, Basak O, Isaacs AM, Westeneng HJ, van den Berg LH, Veldink JH, Schlegel DK, Pasterkamp RJ. Molecular pathology, developmental changes and synaptic dysfunction in (pre-) symptomatic human C9ORF72-ALS/FTD cerebral organoids. Acta Neuropathol Commun 2024; 12:152. [PMID: 39289761 PMCID: PMC11409520 DOI: 10.1186/s40478-024-01857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
A hexanucleotide repeat expansion (HRE) in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Human brain imaging and experimental studies indicate early changes in brain structure and connectivity in C9-ALS/FTD, even before symptom onset. Because these early disease phenotypes remain incompletely understood, we generated iPSC-derived cerebral organoid models from C9-ALS/FTD patients, presymptomatic C9ORF72-HRE (C9-HRE) carriers, and controls. Our work revealed the presence of all three C9-HRE-related molecular pathologies and developmental stage-dependent size phenotypes in cerebral organoids from C9-ALS/FTD patients. In addition, single-cell RNA sequencing identified changes in cell type abundance and distribution in C9-ALS/FTD organoids, including a reduction in the number of deep layer cortical neurons and the distribution of neural progenitors. Further, molecular and cellular analyses and patch-clamp electrophysiology detected various changes in synapse structure and function. Intriguingly, organoids from all presymptomatic C9-HRE carriers displayed C9-HRE molecular pathology, whereas the extent to which more downstream cellular defects, as found in C9-ALS/FTD models, were detected varied for the different presymptomatic C9-HRE cases. Together, these results unveil early changes in 3D human brain tissue organization and synaptic connectivity in C9-ALS/FTD that likely constitute initial pathologies crucial for understanding disease onset and the design of therapeutic strategies.
Collapse
Affiliation(s)
- Astrid T van der Geest
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Channa E Jakobs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tijana Ljubikj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan F M Huffels
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Renata Vieira de Sá
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Daan H Rutten
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marthe Kaal
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maria M Zwartkruis
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Ewout J N Groen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henk-Jan Westeneng
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Domino K Schlegel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
19
|
Zelina P, de Ruiter AA, Kolsteeg C, van Ginneken I, Vos HR, Supiot LF, Burgering BMT, Meye FJ, Veldink JH, van den Berg LH, Pasterkamp RJ. ALS-associated C21ORF2 variant disrupts DNA damage repair, mitochondrial metabolism, neuronal excitability and NEK1 levels in human motor neurons. Acta Neuropathol Commun 2024; 12:144. [PMID: 39227882 PMCID: PMC11373222 DOI: 10.1186/s40478-024-01852-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease leading to motor neuron loss. Currently mutations in > 40 genes have been linked to ALS, but the contribution of many genes and genetic mutations to the ALS pathogenic process remains poorly understood. Therefore, we first performed comparative interactome analyses of five recently discovered ALS-associated proteins (C21ORF2, KIF5A, NEK1, TBK1, and TUBA4A) which highlighted many novel binding partners, and both unique and shared interactors. The analysis further identified C21ORF2 as a strongly connected protein. The role of C21ORF2 in neurons and in the nervous system, and of ALS-associated C21ORF2 variants is largely unknown. Therefore, we combined human iPSC-derived motor neurons with other models and different molecular cell biological approaches to characterize the potential pathogenic effects of C21ORF2 mutations in ALS. First, our data show C21ORF2 expression in ALS-relevant mouse and human neurons, such as spinal and cortical motor neurons. Further, the prominent ALS-associated variant C21ORF2-V58L caused increased apoptosis in mouse neurons and movement defects in zebrafish embryos. iPSC-derived motor neurons from C21ORF2-V58L-ALS patients, but not isogenic controls, show increased apoptosis, and changes in DNA damage response, mitochondria and neuronal excitability. In addition, C21ORF2-V58L induced post-transcriptional downregulation of NEK1, an ALS-associated protein implicated in apoptosis and DDR. In all, our study defines the pathogenic molecular and cellular effects of ALS-associated C21ORF2 mutations and implicates impaired post-transcriptional regulation of NEK1 downstream of mutant C21ORF72 in ALS.
Collapse
Affiliation(s)
- Pavol Zelina
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Anna Aster de Ruiter
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Christy Kolsteeg
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Ilona van Ginneken
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Harmjan R Vos
- Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Laura F Supiot
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
20
|
Fang T, Pacut P, Bose A, Sun Y, Gao J, Sivakumar S, Bloom B, Nascimento Andrade EI, Trombetta B, Ghasemi M. Clinical and genetic factors affecting diagnostic timeline of amyotrophic lateral sclerosis: a 15-year retrospective study. Neurol Res 2024; 46:859-867. [PMID: 38825034 DOI: 10.1080/01616412.2024.2362578] [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: 09/28/2023] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
OBJECTIVES Amyotrophic Lateral Sclerosis (ALS) diagnosis can take 10-16 months from symptom onset, leading to delays in treatment and patient counselling. We studied the impact of clinical and genetic risk factors on the diagnostic timeline of ALS. METHODS Baseline characteristics, family history, gene testing, onset location, time from symptom onset to diagnosis, and time from first doctor visit to suspected ALS was collected. We used multiple regression to assess the interaction of these factors on ALS diagnostic timeline. We analysed a subgroup of patients with genetic testing and compared positive or negative tests, sporadic or familial and ALS-related genes to time for diagnosis. RESULTS Four hundred and forty-eight patients diagnosed with ALS at the University of Massachusetts Chan Medical Center between January 2007 and December 2021 were analysed. The median time to ALS diagnosis was 12 months and remained unchanged from 2007 to 2021 (p = 0.20). Diagnosis was delayed in patients with sporadic compared with familial ALS (mean months [standard deviation], 16.5[13.5] and 11.2[8.5], p < 0.001); cognitive onset (41[21.26]) had longer time to diagnosis than bulbar (11.9[8.2]), limb (15.9[13.2]), respiratory (19.7[13.9]) and ALS with multiple onset locations (20.77[15.71], p < 0.001). One hundred and thirty-four patients had gene testing and 32 tested positive (23.8%). Gene testing (p = 0.23), a positive genetic test (p = 0.16), different ALS genes (p = 0.25) and sporadic (p = 0.92) or familial (p = 0.85) ALS testing positive for ALS genes did not influence time to diagnosis. DISCUSSION Time for ALS diagnosis remained unchanged from 2007 to 2021, bulbar-onset and familial ALS made for faster diagnosis.
Collapse
Affiliation(s)
- Ton Fang
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Peter Pacut
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Abigail Bose
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yuyao Sun
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Jeff Gao
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shravan Sivakumar
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Brooke Bloom
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Bianca Trombetta
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Neurology, Lahey Hospital and Medical Center, Burlington, MA, USA
| |
Collapse
|
21
|
Calma AD, van den Bos M, Pavey N, Santos Silva C, Menon P, Vucic S. Physiological Biomarkers of Upper Motor Neuron Dysfunction in ALS. Brain Sci 2024; 14:760. [PMID: 39199454 PMCID: PMC11352893 DOI: 10.3390/brainsci14080760] [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: 06/03/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
Upper motor neuron (UMN) dysfunction is an important feature of amyotrophic lateral sclerosis (ALS) for the diagnosis and understanding of pathogenesis. The identification of UMN signs forms the basis of ALS diagnosis, although may be difficult to discern, especially in the setting of severe muscle weakness. Transcranial magnetic stimulation (TMS) techniques have yielded objective physiological biomarkers of UMN dysfunction in ALS, enabling the interrogation of cortical and subcortical neuronal networks with diagnostic, pathophysiological, and prognostic implications. Transcranial magnetic stimulation techniques have provided pertinent pathogenic insights and yielded novel diagnostic and prognostic biomarkers. Cortical hyperexcitability, as heralded by a reduction in short interval intracortical inhibition (SICI) and an increase in short interval intracortical facilitation (SICF), has been associated with lower motor neuron degeneration, patterns of disease evolution, as well as the development of specific ALS clinical features including the split hand phenomenon. Reduction in SICI has also emerged as a potential diagnostic aid in ALS. More recently, physiological distinct inhibitory and facilitatory cortical interneuronal circuits have been identified, which have been shown to contribute to ALS pathogenesis. The triple stimulation technique (TST) was shown to enhance the diagnostic utility of conventional TMS measures in detecting UMN dysfunction. Resting-state EEG is a novel neurophysiological technique developed for directly interrogating cortical neuronal networks in ALS, that have yielded potentially useful physiological biomarkers of UMN dysfunction. The present review discusses physiological biomarkers of UMN dysfunction in ALS, encompassing conventional and novel TMS techniques developed to interrogate the functional integrity of the corticomotoneuronal system, focusing on pathogenic, diagnostic, and prognostic utility.
Collapse
Affiliation(s)
- Aicee Dawn Calma
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Mehdi van den Bos
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Nathan Pavey
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Cláudia Santos Silva
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
- Department of Neurosciences and Mental Health, Unidade Local de Saúde de Santa Maria, 1649-028 Lisbon, Portugal
- Faculdade de Medicina-Instituto de Medicina Molecular, Centro de Estudos Egas Moniz, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Parvathi Menon
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney 2139, Australia (C.S.S.)
| |
Collapse
|
22
|
Hardiman O. Amyotrophic lateral sclerosis: a lesson in translation. Lancet Neurol 2024; 23:651-653. [PMID: 38782014 DOI: 10.1016/s1474-4422(24)00223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Orla Hardiman
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
23
|
Dharmadasa T, Pavey N, Tu S, Menon P, Huynh W, Mahoney CJ, Timmins HC, Higashihara M, van den Bos M, Shibuya K, Kuwabara S, Grosskreutz J, Kiernan MC, Vucic S. Novel approaches to assessing upper motor neuron dysfunction in motor neuron disease/amyotrophic lateral sclerosis: IFCN handbook chapter. Clin Neurophysiol 2024; 163:68-89. [PMID: 38705104 DOI: 10.1016/j.clinph.2024.04.010] [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: 10/01/2023] [Revised: 02/08/2024] [Accepted: 04/14/2024] [Indexed: 05/07/2024]
Abstract
Identifying upper motor neuron (UMN) dysfunction is fundamental to the diagnosis and understanding of disease pathogenesis in motor neuron disease (MND). The clinical assessment of UMN dysfunction may be difficult, particularly in the setting of severe muscle weakness. From a physiological perspective, transcranial magnetic stimulation (TMS) techniques provide objective biomarkers of UMN dysfunction in MND and may also be useful to interrogate cortical and network function. Single, paired- and triple pulse TMS techniques have yielded novel diagnostic and prognostic biomarkers in MND, and have provided important pathogenic insights, particularly pertaining to site of disease onset. Cortical hyperexcitability, as heralded by reduced short interval intracortical inhibition (SICI) and increased short interval intracortical facilitation, has been associated with the onset of lower motor neuron degeneration, along with patterns of disease spread, development of specific clinical features such as the split hand phenomenon, and may provide an indication about the rate of disease progression. Additionally, reduction of SICI has emerged as a potential diagnostic aid in MND. The triple stimulation technique (TST) was shown to enhance the diagnostic utility of conventional TMS measures in detecting UMN dysfunction in MND. Separately, sophisticated brain imaging techniques have uncovered novel biomarkers of neurodegeneration that have bene associated with progression. The present review will discuss the utility of TMS and brain neuroimaging derived biomarkers of UMN dysfunction in MND, focusing on recently developed TMS techniques and advanced neuroimaging modalities that interrogate structural and functional integrity of the corticomotoneuronal system, with an emphasis on pathogenic, diagnostic, and prognostic utility.
Collapse
Affiliation(s)
- Thanuja Dharmadasa
- Department of Neurology, The Royal Melbourne Hospital City Campus, Parkville, Victoria, Australia
| | - Nathan Pavey
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia
| | - Sicong Tu
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Parvathi Menon
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia
| | - William Huynh
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Colin J Mahoney
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Hannah C Timmins
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Mehdi van den Bos
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia
| | - Kazumoto Shibuya
- Neurology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Satoshi Kuwabara
- Neurology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Julian Grosskreutz
- Precision Neurology, Excellence Cluster Precision Medicine in Inflammation, University of Lübeck, University Hospital Schleswig-Holstein Campus, Lübeck, Germany
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, and Department of Neurology, Royal Prince Alfred Hospital, Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney, Australia.
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
McFarlane R, Heverin M, Walsh C, Hardiman O. Irish Amyotrophic Lateral Sclerosis Incidence: Age, Period, and Cohort Effects Using a Partial Least Squares Regression Model. Neurology 2024; 102:e209391. [PMID: 38728654 DOI: 10.1212/wnl.0000000000209391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES To investigate the underlying reasons for variability in the incidence rate of amyotrophic lateral sclerosis (ALS) within the Irish population between the years 1996 and 2021. METHODS The Irish ALS register was used to calculate the incidence and to subsequently extract age at diagnosis (age), year of diagnosis (period), and date of birth (cohort) for all incident patients within the study period (n = 2,771). An age-period-cohort (APC) model using partial least squares regression was constructed to examine each component separately and their respective contribution to the incidence while minimizing the well-known identifiability problem of APC effects. A dummy regression model consisting of 5 periods, 19 cohorts, and 16 age groups was used to examine nonlinear relationships within the data over time. The CIs for each of these were estimated using the jackknife method. RESULTS The nonlinear model achieved R2 of 99.43% with 2-component extraction. Age variation was evident with those in the ages 65-79 years contributing significantly to the incidence (βmax = 0.0746, SE = 0.000410, CI 0.00665-0.00826). However, those aged 25-60 years contributed significantly less (βmin = -0.00393, SE = 0.000291, CI -0.00454 to -0.00340). Each successive period showed an increase in the regression model coefficient suggesting an increasing incidence over time, independent of the other factors examined-an increase of β from -0.00489 (SE = 0.000264, CI -0.00541 to -0.00437) to 0.00973 (SE = 0.000418, CI 0.0105-0.00891). A cohort effect was demonstrated showing that the contribution of those born between 1927 and 1951 contributed to a significantly greater degree than the other birth cohorts (βmax = 0.00577, SE = 0.000432, CI 0.00493-0.00662). DISCUSSION Using the Irish population-based ALS Register, robust age, period, and cohort effects can be identified. The age effect may be accounted for by demographic shifts within the population. Changes in disease categorization, competing risks of death, and improved surveillance may account for period effects. The cohort effect may reflect lifestyle and environmental factors associated with the challenging economic circumstances in Ireland between 1927 and 1951. Age-period-cohort studies can help to account for changes in disease incidence and prevalence, providing additional insights into likely demographic and environmental factors that influence population-based disease risk.
Collapse
Affiliation(s)
- Robert McFarlane
- From the Academic Unit of Neurology (R.M., M.H., O.H.), and Department of Biostatistics (C.W.), Trinity College Dublin, Ireland
| | - Mark Heverin
- From the Academic Unit of Neurology (R.M., M.H., O.H.), and Department of Biostatistics (C.W.), Trinity College Dublin, Ireland
| | - Cathal Walsh
- From the Academic Unit of Neurology (R.M., M.H., O.H.), and Department of Biostatistics (C.W.), Trinity College Dublin, Ireland
| | - Orla Hardiman
- From the Academic Unit of Neurology (R.M., M.H., O.H.), and Department of Biostatistics (C.W.), Trinity College Dublin, Ireland
| |
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
|
Evans LJ, O'Brien D, Shaw PJ. Current neuroprotective therapies and future prospects for motor neuron disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:327-384. [PMID: 38802178 DOI: 10.1016/bs.irn.2024.04.003] [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
Four medications with neuroprotective disease-modifying effects are now in use for motor neuron disease (MND). With FDA approvals for tofersen, relyvrio and edaravone in just the past year, 2022 ended a quarter of a century when riluzole was the sole such drug to offer to patients. The acceleration of approvals may mean we are witnessing the beginning of a step-change in how MND can be treated. Improvements in understanding underlying disease biology has led to more therapies being developed to target specific and multiple disease mechanisms. Consideration for how the pipeline of new therapeutic agents coming through in clinical and preclinical development can be more effectively evaluated with biomarkers, advances in patient stratification and clinical trial design pave the way for more successful translation for this archetypal complex neurodegenerative disease. While it must be cautioned that only slowed rates of progression have so far been demonstrated, pre-empting rapid neurodegeneration by using neurofilament biomarkers to signal when to treat, as is currently being trialled with tofersen, may be more effective for patients with known genetic predisposition to MND. Early intervention with personalized medicines could mean that for some patients at least, in future we may be able to substantially treat what is considered by many to be one of the most distressing diseases in medicine.
Collapse
Affiliation(s)
- Laura J Evans
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - David O'Brien
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Pamela J Shaw
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
Dai T, Lou J, Kong D, Li J, Ren Q, Chen Y, Sun S, Yun Y, Sun X, Yang Y, Shao K, Li W, Zhao Y, Meng X, Yan C, Lin P, Liu S. Choroid plexus enlargement in amyotrophic lateral sclerosis patients and its correlation with clinical disability and blood-CSF barrier permeability. Fluids Barriers CNS 2024; 21:36. [PMID: 38632611 PMCID: PMC11025206 DOI: 10.1186/s12987-024-00536-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Using in vivo neuroimaging techniques, growing evidence has demonstrated that the choroid plexus (CP) volume is enlarged in patients with several neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. However, although animal and postmortem findings suggest that CP abnormalities are likely important pathological mechanisms underlying amyotrophic lateral sclerosis (ALS), the third most common neurodegenerative disease, no available study has been conducted to thoroughly assess CP abnormalities and their clinical relevance in vivo in ALS patients to date. Thus, we aimed to determine whether in vivo CP enlargement may occur in ALS patients. We also aimed to identify the relationships of CP volume with clinical disabilities and blood-CSF barrier (BCSFB) permeability in ALS patients. METHODS In this retrospective study, based on structural MRI data, CP volume was assessed using a Gaussian mixture model and underwent further manual correction in 155 ALS patients and 105 age- and sex-matched HCs from October 2021 to April 2023. The ALS Functional Rating Scale-Revised (ALSFRS-R) was used to assess clinical disability. The CSF/serum albumin quotient (Qalb) was used to assess BCSFB permeability. Moreover, all the ALS patients completed genetic testing, and according to genetic testing, the ALS patients were further divided into genetic ALS subgroup and sporadic ALS subgroup. RESULTS We found that compared with HCs, ALS patients had a significantly higher CP volume (p < 0.001). Moreover, compared with HCs, CP volume was significantly increased in both ALS patients with and without known genetic mutations after family-wise error correction (p = 0.006 and p < 0.001, respectively), while there were no significant differences between the two ALS groups. Furthermore, the CP volume was significantly correlated with the ALSFRS-r score (r = -0.226; p = 0.005) and the Qalb (r = 0.479; p < 0.001) in ALS patients. CONCLUSION Our study first demonstrates CP enlargement in vivo in ALS patients, and continues to suggest an important pathogenetic role for CP abnormalities in ALS. Moreover, assessing CP volume is likely a noninvasive and easy-to-implement approach for screening BCSFB dysfunction in ALS patients.
Collapse
Affiliation(s)
- Tingjun Dai
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
| | - Jianwei Lou
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
| | - Deyuan Kong
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jinyu Li
- Department of Neurology, Xiamen Branch, Zhongshan Hospital, Fudan University, 361015, Xiamen, China
| | - Qingguo Ren
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Yujing Chen
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
| | - Sujuan Sun
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
| | - Yan Yun
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Xiaohan Sun
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
| | - Yiru Yang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kai Shao
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wei Li
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
| | - Yuying Zhao
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
| | - Xiangshui Meng
- Department of Radiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Chuanzhu Yan
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Pengfei Lin
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China.
| | - Shuangwu Liu
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, West Wenhua Street No.107, 250012, Jinan, China.
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
35
|
Libonati L, Cambieri C, Colavito D, Moret F, D'Andrea E, Del Giudice E, Leon A, Inghilleri M, Ceccanti M. Genetics screening in an Italian cohort of patients with Amyotrophic Lateral Sclerosis: the importance of early testing and its implication. J Neurol 2024; 271:1921-1936. [PMID: 38112783 DOI: 10.1007/s00415-023-12142-x] [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: 09/06/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease with an elusive etiology. While environmental factors have been considered, familial ALS cases have raised the possibility of genetic involvement. This genetic connection is increasingly evident, even in patients with sporadic ALS. We allowed access to the genetic test to all patients attending our clinic to identify the prevalence and the role of genetic variants in the development of the disease and to identify patients with potentially treatable forms of the disease. MATERIALS AND METHODS 194 patients with probable or definite ALS, were enrolled. A comprehensive genetic testing was performed, including sequencing all exons of the SOD1 gene and testing for hexanucleotide intronic repeat expansions (G4C2) in the C9orf72 gene using fluorescent repeat-primed PCR (RP-PCR). Whole Exome NGS Sequencing (WES) was performed, followed by an in silico multigene panel targeting neuromuscular diseases, spastic paraplegia, and motor distal neuropathies. We conducted statistical analyses to compare different patient groups. RESULTS Clinically significant pathogenetic variants were detected in 14.43% of cases. The highest prevalence of pathogenetic variants was observed in fALS patients, but a substantial proportion of sALS patients also displayed at least one variant, either pathogenetic or of uncertain significance (VUS). The most observed pathogenetic variant was the expansion of the C9orf72 gene, which was associated with a shorter survival. SOD1 variants were found in 1.6% of fALS and 2.5% of sALS patients. DISCUSSION The study reveals a significant number of ALS patients carrying pathogenic or likely pathogenic variants, with a higher prevalence in familial ALS cases. The expansion of the C9orf72 gene emerges as the most common genetic cause of ALS, affecting familial and sporadic cases. Additionally, SOD1 variants are detected at an unexpectedly higher rate, even in patients without a familial history of ALS, underscoring the crucial role of genetic testing in treatment decisions and potential participation in clinical trials. We also investigated variants in genes such as TARDBP, FUS, NEK1, TBK1, and DNAJC7, shedding light on their potential involvement in ALS. These findings underscore the complexity of interpreting variants of uncertain significance (VUS) and their ethical implications in patient communication and genetic counseling for patients' relatives. CONCLUSION This study emphasizes the diverse genetic basis of ALS and advocates for integrating comprehensive genetic testing into diagnostic protocols. The evolving landscape of genetic therapies requires identifying all eligible patients transcending traditional familial boundaries. The presence of VUS highlights the multifaceted nature of ALS genetics, prompting further exploration of complex interactions among genetic variants, environmental factors, and disease development.
Collapse
Affiliation(s)
- Laura Libonati
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, Viale Dell'Università 30, 00185, Rome, Italy.
| | - Chiara Cambieri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, Viale Dell'Università 30, 00185, Rome, Italy
| | - Davide Colavito
- R & I Genetics, C.So Stati Uniti 4int.F, 35127, Padua, Italy
| | - Federica Moret
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, Viale Dell'Università 30, 00185, Rome, Italy
| | - Edoardo D'Andrea
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, Viale Dell'Università 30, 00185, Rome, Italy
| | | | - Alberta Leon
- R & I Genetics, C.So Stati Uniti 4int.F, 35127, Padua, Italy
| | - Maurizio Inghilleri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, Viale Dell'Università 30, 00185, Rome, Italy
| | - Marco Ceccanti
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, Viale Dell'Università 30, 00185, Rome, Italy
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
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
|
39
|
Shi J, Wang Z, Yi M, Xie S, Zhang X, Tao D, Liu Y, Yang Y. Evidence based on Mendelian randomization and colocalization analysis strengthens causal relationships between structural changes in specific brain regions and risk of amyotrophic lateral sclerosis. Front Neurosci 2024; 18:1333782. [PMID: 38505770 PMCID: PMC10948422 DOI: 10.3389/fnins.2024.1333782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the degeneration of motor neurons in the brain and spinal cord with a poor prognosis. Previous studies have observed cognitive decline and changes in brain morphometry in ALS patients. However, it remains unclear whether the brain structural alterations contribute to the risk of ALS. In this study, we conducted a bidirectional two-sample Mendelian randomization (MR) and colocalization analysis to investigate this causal relationship. Methods Summary data of genome-wide association study were obtained for ALS and the brain structures, including surface area (SA), thickness and volume of subcortical structures. Inverse-variance weighted (IVW) method was used as the main estimate approach. Sensitivity analysis was conducted detect heterogeneity and pleiotropy. Colocalization analysis was performed to calculate the posterior probability of causal variation and identify the common genes. Results In the forward MR analysis, we found positive associations between the SA in four cortical regions (lingual, parahippocampal, pericalcarine, and middle temporal) and the risk of ALS. Additionally, decreased thickness in nine cortical regions (caudal anterior cingulate, frontal pole, fusiform, inferior temporal, lateral occipital, lateral orbitofrontal, pars orbitalis, pars triangularis, and pericalcarine) was significantly associated with a higher risk of ALS. In the reverse MR analysis, genetically predicted ALS was associated with reduced thickness in the bankssts and increased thickness in the caudal middle frontal, inferior parietal, medial orbitofrontal, and superior temporal regions. Colocalization analysis revealed the presence of shared causal variants between the two traits. Conclusion Our results suggest that altered brain morphometry in individuals with high ALS risk may be genetically mediated. The causal associations of widespread multifocal extra-motor atrophy in frontal and temporal lobes with ALS risk support the notion of a continuum between ALS and frontotemporal dementia. These findings enhance our understanding of the cortical structural patterns in ALS and shed light on potentially viable therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yuan Yang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Jagaraj CJ, Shadfar S, Kashani SA, Saravanabavan S, Farzana F, Atkin JD. Molecular hallmarks of ageing in amyotrophic lateral sclerosis. Cell Mol Life Sci 2024; 81:111. [PMID: 38430277 PMCID: PMC10908642 DOI: 10.1007/s00018-024-05164-9] [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: 12/05/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, severely debilitating and rapidly progressing disorder affecting motor neurons in the brain, brainstem, and spinal cord. Unfortunately, there are few effective treatments, thus there remains a critical need to find novel interventions that can mitigate against its effects. Whilst the aetiology of ALS remains unclear, ageing is the major risk factor. Ageing is a slowly progressive process marked by functional decline of an organism over its lifespan. However, it remains unclear how ageing promotes the risk of ALS. At the molecular and cellular level there are specific hallmarks characteristic of normal ageing. These hallmarks are highly inter-related and overlap significantly with each other. Moreover, whilst ageing is a normal process, there are striking similarities at the molecular level between these factors and neurodegeneration in ALS. Nine ageing hallmarks were originally proposed: genomic instability, loss of telomeres, senescence, epigenetic modifications, dysregulated nutrient sensing, loss of proteostasis, mitochondrial dysfunction, stem cell exhaustion, and altered inter-cellular communication. However, these were recently (2023) expanded to include dysregulation of autophagy, inflammation and dysbiosis. Hence, given the latest updates to these hallmarks, and their close association to disease processes in ALS, a new examination of their relationship to pathophysiology is warranted. In this review, we describe possible mechanisms by which normal ageing impacts on neurodegenerative mechanisms implicated in ALS, and new therapeutic interventions that may arise from this.
Collapse
Affiliation(s)
- Cyril Jones Jagaraj
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sina Shadfar
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sara Assar Kashani
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sayanthooran Saravanabavan
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Fabiha Farzana
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Julie D Atkin
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia.
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
41
|
He D, Liu Y, Dong S, Shen D, Yang X, Hao M, Yin X, He X, Li Y, Wang Y, Liu M, Wang J, Chen X, Cui L. The prognostic value of systematic genetic screening in amyotrophic lateral sclerosis patients. J Neurol 2024; 271:1385-1396. [PMID: 37980296 DOI: 10.1007/s00415-023-12079-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/20/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with complex genetic architecture. Emerging evidence has indicated comorbidity between ALS and autoimmune conditions, suggesting a potential shared genetic basis. The objective of this study is to assess the prognostic value of systematic screening for rare deleterious mutations in genes associated with ALS and aberrant inflammatory responses. METHODS A discovery cohort of 494 patients and a validation cohort of 69 patients were analyzed in this study, with population-matched healthy subjects (n = 4961) served as controls. Whole exome sequencing (WES) was performed to identify rare deleterious variants in 50 ALS genes and 1177 genes associated with abnormal inflammatory responses. Genotype-phenotype correlation was assessed, and an integrative prognostic model incorporating genetic and clinical factors was constructed. RESULTS In the discovery cohort, 8.1% of patients carried confirmed ALS variants, and an additional 15.2% of patients carried novel ALS variants. Gene burden analysis revealed 303 immune-implicated genes with enriched rare variants, and 13.4% of patients harbored rare deleterious variants in these genes. Patients with ALS variants exhibited a more rapid disease progression (HR 2.87 [95% CI 2.03-4.07], p < 0.0001), while no significant effect was observed for immune-implicated variants. The nomogram model incorporating genetic and clinical information demonstrated improved accuracy in predicting disease outcomes (C-index, 0.749). CONCLUSION Our findings enhance the comprehension of the genetic basis of ALS within the Chinese population. It also appears that rare deleterious mutations occurring in immune-implicated genes exert minimal influence on the clinical trajectories of ALS patients.
Collapse
Affiliation(s)
- Di He
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Yining Liu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Siqi Dong
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Dongchao Shen
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Xunzhe Yang
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Meng Hao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Xianhong Yin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Xinyi He
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yi Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yi Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Mingsheng Liu
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China.
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China.
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, National Center for Neurological Disorders, Fudan University, Shanghai, China.
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China.
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Dongcheng District, Beijing, China.
| |
Collapse
|
42
|
Marrie RA, Maxwell CJ, Rotstein DL, Tsai CC, Tremlett H. Prodromes in demyelinating disorders, amyotrophic lateral sclerosis, Parkinson disease, and Alzheimer's dementia. Rev Neurol (Paris) 2024; 180:125-140. [PMID: 37567819 DOI: 10.1016/j.neurol.2023.07.002] [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: 05/29/2023] [Revised: 06/24/2023] [Accepted: 07/03/2023] [Indexed: 08/13/2023]
Abstract
A prodrome is an early set of symptoms, which indicates the onset of a disease; these symptoms are often non-specific. Prodromal phases are now recognized in multiple central nervous system diseases. The depth of understanding of the prodromal phase varies across diseases, being more nascent for multiple sclerosis for example, than for Parkinson disease or Alzheimer's disease. Key challenges when identifying the prodromal phase of a disease include the lack of specificity of prodromal symptoms, and consequent need for accessible and informative biomarkers. Further, heterogeneity of the prodromal phase may be influenced by age, sex, genetics and other poorly understood factors. Nonetheless, recognition that an individual is in the prodromal phase of disease offers the opportunity for earlier diagnosis and with it the opportunity for earlier intervention.
Collapse
Affiliation(s)
- R A Marrie
- Departments of Internal Medicine and Community Health Sciences, Rady Faculty of Health Sciences, Max-Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - C J Maxwell
- Schools of Pharmacy and Public Health Sciences, University of Waterloo, Waterloo, Ontario, Canada; ICES, Toronto, Ontario, Canada
| | - D L Rotstein
- Department of Medicine, University of Toronto, 6, Queen's Park Crescent West, 3rd floor, M5S 3H2 Toronto, Ontario, Canada; Saint-Michael's Hospital, 30, Bond Street, M5B 1W8 Toronto, Ontario, Canada
| | - C-C Tsai
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - H Tremlett
- Faculty of Medicine (Neurology), University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
43
|
Lee A, Henderson R, Aylward J, McCombe P. Gut Symptoms, Gut Dysbiosis and Gut-Derived Toxins in ALS. Int J Mol Sci 2024; 25:1871. [PMID: 38339149 PMCID: PMC10856138 DOI: 10.3390/ijms25031871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Many pathogenetic mechanisms have been proposed for amyotrophic lateral sclerosis (ALS). Recently, there have been emerging suggestions of a possible role for the gut microbiota. Gut microbiota have a range of functions and could influence ALS by several mechanisms. Here, we review the possible role of gut-derived neurotoxins/excitotoxins. We review the evidence of gut symptoms and gut dysbiosis in ALS. We then examine a possible role for gut-derived toxins by reviewing the evidence that these molecules are toxic to the central nervous system, evidence of their association with ALS, the existence of biochemical pathways by which these molecules could be produced by the gut microbiota and existence of mechanisms of transport from the gut to the blood and brain. We then present evidence that there are increased levels of these toxins in the blood of some ALS patients. We review the effects of therapies that attempt to alter the gut microbiota or ameliorate the biochemical effects of gut toxins. It is possible that gut dysbiosis contributes to elevated levels of toxins and that these could potentially contribute to ALS pathogenesis, but more work is required.
Collapse
Affiliation(s)
- Aven Lee
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (R.H.); (P.M.)
| | - Robert Henderson
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (R.H.); (P.M.)
- Department of Neurology, Royal Brisbane & Women’s Hospital, Brisbane, QLD 4029, Australia
- Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia;
| | - James Aylward
- Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia;
| | - Pamela McCombe
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; (R.H.); (P.M.)
- Department of Neurology, Royal Brisbane & Women’s Hospital, Brisbane, QLD 4029, Australia
- Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia;
| |
Collapse
|
44
|
Borrego-Hernández D, Vázquez-Costa JF, Domínguez-Rubio R, Expósito-Blázquez L, Aller E, Padró-Miquel A, García-Casanova P, Colomina MJ, Martín-Arriscado C, Osta R, Cordero-Vázquez P, Esteban-Pérez J, Povedano-Panadés M, García-Redondo A. Intermediate Repeat Expansion in the ATXN2 Gene as a Risk Factor in the ALS and FTD Spanish Population. Biomedicines 2024; 12:356. [PMID: 38397958 PMCID: PMC10886453 DOI: 10.3390/biomedicines12020356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Intermediate CAG expansions in the gene ataxin-2 (ATXN2) are a known risk factor for ALS, but little is known about their role in FTD risk. Moreover, their contribution to the risk and phenotype of patients might vary in populations with different genetic backgrounds. The aim of this study was to assess the relationship of intermediate CAG expansions in ATXN2 with the risk and phenotype of ALS and FTD in the Spanish population. Repeat-primed PCR was performed in 620 ALS and 137 FTD patients in three referral centers in Spain to determine the exact number of CAG repeats. In our cohort, ≥27 CAG repeats in ATXN2 were associated with a higher risk of developing ALS (odds ratio [OR] = 2.666 [1.471-4.882]; p = 0.0013) but not FTD (odds ratio [OR] = 1.446 [0.558-3.574]; p = 0.44). Moreover, ALS patients with ≥27 CAG repeats in ATXN2 showed a shorter survival rate compared to those with <27 repeats (hazard ratio [HR] 1.74 [1.18, 2.56], p = 0.005), more frequent limb onset (odds ratio [OR] = 2.34 [1.093-4.936]; p = 0.028) and a family history of ALS (odds ratio [OR] = 2.538 [1.375-4.634]; p = 0.002). Intermediate CAG expansions of ≥27 repeats in ATXN2 are associated with ALS risk but not with FTD in the Spanish population. ALS patients carrying an intermediate expansion in ATXN2 show more frequent limb onset but a worse prognosis than those without expansions. In patients carrying C9orf72 expansions, the intermediate ATXN2 expansion might increase the penetrance and modify the phenotype.
Collapse
Affiliation(s)
- Daniel Borrego-Hernández
- ALS Research Laboratory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (L.E.-B.); (P.C.-V.); (J.E.-P.); (A.G.-R.)
| | - Juan Francisco Vázquez-Costa
- Neuromuscular Unit, ERN-NMD Group, Department of Neurology, Hospital Universitario y Politécnico La Fe, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.F.V.-C.); (P.G.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain;
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Raúl Domínguez-Rubio
- Motoneuron Functional Unit, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.D.-R.); (M.P.-P.)
| | - Laura Expósito-Blázquez
- ALS Research Laboratory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (L.E.-B.); (P.C.-V.); (J.E.-P.); (A.G.-R.)
| | - Elena Aller
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain;
- Genetics Department, Hospital Universitario y Politécnico La Fe, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Ariadna Padró-Miquel
- Genetics Laboratory (LCTMS), Bellvitge University Hospital-IDIBELL, 08908 L’Hospitalet de Llobregat, Spain;
| | - Pilar García-Casanova
- Neuromuscular Unit, ERN-NMD Group, Department of Neurology, Hospital Universitario y Politécnico La Fe, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.F.V.-C.); (P.G.-C.)
| | - María J. Colomina
- Anesthesia Service Unit, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain;
| | | | - Rosario Osta
- Laboratório de Genética e Biotecnologia (LAGENBIO), Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Aragon Institute for Health Research (IIS Aragon), Zaragoza University, 50013 Zaragoza, Spain;
| | - Pilar Cordero-Vázquez
- ALS Research Laboratory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (L.E.-B.); (P.C.-V.); (J.E.-P.); (A.G.-R.)
| | - Jesús Esteban-Pérez
- ALS Research Laboratory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (L.E.-B.); (P.C.-V.); (J.E.-P.); (A.G.-R.)
| | - Mónica Povedano-Panadés
- Motoneuron Functional Unit, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.D.-R.); (M.P.-P.)
| | - Alberto García-Redondo
- ALS Research Laboratory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (L.E.-B.); (P.C.-V.); (J.E.-P.); (A.G.-R.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain;
| |
Collapse
|
45
|
Vélez-GóMEZ B, Perna A, Vazquez C, Ketzoian C, Lillo P, Godoy-Reyes G, Sáez D, Zaldivar Vaillant T, Gutiérrez Gil JV, Lara-Fernández GE, Povedano M, Heverin M, McFarlane R, Logroscino G, Hardiman O. LAENALS: epidemiological and clinical features of amyotrophic lateral sclerosis in Latin America. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:119-127. [PMID: 37865869 DOI: 10.1080/21678421.2023.2271517] [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: 07/21/2023] [Accepted: 10/05/2023] [Indexed: 10/23/2023]
Abstract
OBJECTIVE The Latin American Epidemiologic study of ALS (LAENALS) aims to gather data on ALS epidemiology, phenotype, and risk factors in Cuba, Chile, and Uruguay, to understand the impact of genetic and environmental factors on ALS. METHODS A harmonized data collection protocol was generated, and a Latin-American Spanish language Register was constructed. Patient data were collected in Uruguay in 2018, in Chile from 2017 to 2019, and in Cuba between 2017 and 2018. Statistical analysis was performed using SPSS 25.0.0 software. Crude cumulative incidence, standardized incidence, and prevalence were calculated in the population aged 15 years and older. RESULTS During 2017-2019, 90 people with ALS from Uruguay (55.6% men), 219 from Chile (54.6% men), and 49 from Cuba (55.1% men) were included. The cumulative crude incidence in 2018 was 1.73/100,000 persons in Uruguay, 1.08 in Chile and 0.195 in Cuba. Crude prevalence in 2018 was 2.19 per 100,000 persons in Uruguay, 1.39 in Chile and 0.55 in Cuba. Mean age at onset was 61.8 ± 11.96 SD years in Uruguay, 61.9 ± 10.4 SD years in Chile, and 60.21 ± 12.45 SD years in Cuba (p = 0.75). Median survival from onset was 32.43 months (21.93 - 42.36) in Uruguay, 24 months (13.5 - 33.5) in Chile, and 29 months (15 - 42.5) in Cuba (p = 0.006). CONCLUSIONS These preliminary data from LAENALS confirm the lower incidence and prevalence of ALS in counties with admixed populations. The LAENALS database is now open to other Latin American countries for harmonized prospective data collection.
Collapse
Affiliation(s)
- Beatriz Vélez-GóMEZ
- Academic Unit of Neurology, Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- Department of Neurology, Hospital Universitario Torrecárdenas, Almería, Spain
| | | | | | | | - Patricia Lillo
- Departamento de Neurología Sur, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Unidad de Neurología, Hospital San Jose, SSMN, Santiago, Chile
| | - Gladys Godoy-Reyes
- Departamento de Neurología Sur, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Servicio de Neurología, Hospital Barros Luco, SSMS, Santiago, Chile
| | - David Sáez
- Departamento de Neurología Sur, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Servicio de Neurología, Hospital Barros Luco, SSMS, Santiago, Chile
| | | | | | | | - Mónica Povedano
- Unidad de Neuromuscular, Servicio de Neurología, Hospital Universitario de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain, and
| | - Mark Heverin
- Academic Unit of Neurology, Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Robert McFarlane
- Academic Unit of Neurology, Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Giancarlo Logroscino
- Center for Neurodegenerative Diseases and the Aging Brain. Fondazione "Card. G. Panico" Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari "Aldo Moro", Bari, Italy
| | - Orla Hardiman
- Academic Unit of Neurology, Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
46
|
Kiernan MC, Farrar MA. Emerging role for sphingolipids in the genetics of amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2024; 95:101-102. [PMID: 38041659 DOI: 10.1136/jnnp-2023-332719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 12/03/2023]
Affiliation(s)
- Matthew C Kiernan
- Bushell Chair of Neurology, Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Neurology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Michelle Anne Farrar
- Neurology, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia
- School of Women's and Children's Health, UNSW Medicine, UNSW, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Moreno-Roco J, del Valle L, Jiménez D, Acosta I, Castillo JL, Dharmadasa T, Kiernan MC, Matamala JM. Diagnostic utility of transcranial magnetic stimulation for neurodegenerative disease: a critical review. Dement Neuropsychol 2024; 17:e20230048. [PMID: 38189033 PMCID: PMC10768644 DOI: 10.1590/1980-5764-dn-2023-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 01/09/2024] Open
Abstract
Neurodegenerative diseases pose significant challenges due to their impact on brain structure, function, and cognition. As life expectancy rises, the prevalence of these disorders is rapidly increasing, resulting in substantial personal, familial, and societal burdens. Efforts have been made to optimize the diagnostic and therapeutic processes, primarily focusing on clinical, cognitive, and imaging characterization. However, the emergence of non-invasive brain stimulation techniques, specifically transcranial magnetic stimulation (TMS), offers unique functional insights and diagnostic potential. TMS allows direct evaluation of brain function, providing valuable information inaccessible through other methods. This review aims to summarize the current and potential diagnostic utility of TMS in investigating neurodegenerative diseases, highlighting its relevance to the field of cognitive neuroscience. The findings presented herein contribute to the growing body of research focused on improving our understanding and management of these debilitating conditions, particularly in regions with limited resources and a pressing need for innovative approaches.
Collapse
Affiliation(s)
- Javier Moreno-Roco
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
| | - Lucía del Valle
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
| | - Daniel Jiménez
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
- Hospital del Salvador, Servicio de Neurología, Santiago, Chile
| | - Ignacio Acosta
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
- Hospital del Salvador, Servicio de Neurología, Santiago, Chile
| | - José Luis Castillo
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
| | - Thanuja Dharmadasa
- University of Melbourne, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- The Royal Melbourne Hospital, Department of Neurology, Parkville, Victoria, Australia
- University of Sydney, Brain and Mind Centre, Sydney, Australia
| | - Matthew C. Kiernan
- University of Sydney, Brain and Mind Centre, Sydney, Australia
- Royal Prince Alfred Hospital, Department of Neurology, Sydney, AustraliaArgento
| | - José Manuel Matamala
- Universidad de Chile, Facultad de Medicina, Laboratorio de Neurología y Neurofisiología Traslacional, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Centro de Investigación Clínica Avanzado (CICA) Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Ciencias Neurológicas Oriente, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Departamento de Neurociencias, Santiago, Chile
- Universidad de Chile, Facultad de Medicina, Instituto de Neurociencia Biomédica (BNI), Santiago, Chile
| |
Collapse
|
48
|
Liu S, Sun X, Ren Q, Chen Y, Dai T, Yang Y, Gong G, Li W, Zhao Y, Meng X, Lin P, Yan C. Glymphatic dysfunction in patients with early-stage amyotrophic lateral sclerosis. Brain 2024; 147:100-108. [PMID: 37584389 DOI: 10.1093/brain/awad274] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/17/2023] [Accepted: 07/30/2023] [Indexed: 08/17/2023] Open
Abstract
Recently, an astrocytic aquaporin 4-dependent drainage system, that is, the glymphatic system, has been identified in the live murine and human brain. Growing evidence suggests that glymphatic function is impaired in patients with several neurodegenerative diseases, including Alzheimer's and Parkinson's disease. As the third most common neurodegenerative disease, although animal studies have indicated that early glymphatic dysfunction is likely an important pathological mechanism underpinning amyotrophic lateral sclerosis (ALS), no available study has been conducted to thoroughly assess glymphatic function in vivo in ALS patients to date, particularly in patients with early-stage ALS. Thus, using diffusion tensor imaging analysis along the perivascular space (ALPS) index, an approximate measure of glymphatic function in vivo, we aimed to explore whether glymphatic function is impaired in patients with patients with early-stage ALS, and the diagnostic performance of the ALPS index in distinguishing between patients with early-stage ALS and healthy subjects. We also aimed to identify the relationships between glymphatic dysfunction and clinical disabilities and sleep problems in patients with early-stage ALS. In this retrospective study, King's Stage 1 ALS patients were defined as patients with early-stage ALS. We enrolled 56 patients with early-stage ALS and 32 age- and sex-matched healthy control subjects. All participants completed clinical screening, sleep assessment and ALPS index analysis. For the sleep assessment, the Pittsburgh Sleep Quality Index, Epworth Sleepiness Scale and polysomnography were used. Compared with healthy control subjects, patients with early-stage ALS had a significantly lower ALPS index after family-wise error correction (P < 0.05). Moreover, receiver operating characteristic analysis showed that the area under the curve for the ALPS index was 0.792 (95% confidence interval 0.700-0.884). Partial correlation analyses showed that the ALPS index was significantly correlated with clinical disability and sleep disturbances in patients with early-stage ALS. Multivariate analysis showed that sleep efficiency (r = 0.419, P = 0.002) and periodic limb movements in sleep index (r = -0.294, P = 0.017) were significant predictive factors of the ALPS index in patients with early-stage ALS. In conclusion, our study continues to support an important role for glymphatic dysfunction in ALS pathology, and we provide additional insights into the early diagnostic value of glymphatic dysfunction and its correlation with sleep disturbances in vivo in patients with early-stage ALS. Moreover, we suggest that early improvement of glymphatic function may be a promising strategy for slowing the neurodegenerative process in ALS. Future studies are needed to explore the diagnostic and therapeutic value of glymphatic dysfunction in individuals with presymptomatic-stage neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuangwu Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266000, China
| | - Xiaohan Sun
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qingguo Ren
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266000, China
| | - Yujing Chen
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tingjun Dai
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yiru Yang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Gaolang Gong
- State Key Laboratory of Cognitive Neuroscience and Learning &IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Wei Li
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yuying Zhao
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiangshui Meng
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266000, China
| | - Pengfei Lin
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chuanzhu Yan
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong 266000, China
| |
Collapse
|
49
|
Vacchiano V, Palombo F, Ormanbekova D, Fiorini C, Fiorentino A, Caporali L, Mastrangelo A, Valentino ML, Capellari S, Liguori R, Carelli V. The genetic puzzle of a SOD1-patient with ocular ptosis and a motor neuron disease: a case report. Front Genet 2023; 14:1322067. [PMID: 38152653 PMCID: PMC10751346 DOI: 10.3389/fgene.2023.1322067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with a complex genetic architecture, showing monogenic, oligogenic, and polygenic inheritance. In this study, we describe the case of a 71 years-old man diagnosed with ALS with atypical clinical features consisting in progressive ocular ptosis and sensorineural deafness. Genetic analyses revealed two heterozygous variants, in the SOD1 (OMIM*147450) and the TBK1 (OMIM*604834) genes respectively, and furthermore mitochondrial DNA (mtDNA) sequencing identified the homoplasmic m.14484T>C variant usually associated with Leber's Hereditary Optic Neuropathy (LHON). We discuss how all these variants may synergically impinge on mitochondrial function, possibly contributing to the pathogenic mechanisms which might ultimately lead to the neurodegenerative process, shaping the clinical ALS phenotype enriched by adjunctive clinical features.
Collapse
Affiliation(s)
- Veria Vacchiano
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Flavia Palombo
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Danara Ormanbekova
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Claudio Fiorini
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Alessia Fiorentino
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Leonardo Caporali
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Andrea Mastrangelo
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Maria Lucia Valentino
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Sabina Capellari
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Valerio Carelli
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
50
|
Violi JP, Pu L, Pravadali-Cekic S, Bishop DP, Phillips CR, Rodgers KJ. Effects of the Toxic Non-Protein Amino Acid β-Methylamino-L-Alanine (BMAA) on Intracellular Amino Acid Levels in Neuroblastoma Cells. Toxins (Basel) 2023; 15:647. [PMID: 37999510 PMCID: PMC10674354 DOI: 10.3390/toxins15110647] [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: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
The cyanobacterial non-protein amino acid (AA) β-Methylamino-L-alanine (BMAA) is considered to be a neurotoxin. BMAA caused histopathological changes in brains and spinal cords of primates consistent with some of those seen in early motor neuron disease; however, supplementation with L-serine protected against some of those changes. We examined the impact of BMAA on AA concentrations in human neuroblastoma cells in vitro. Cells were treated with 1000 µM BMAA and intracellular free AA concentrations in treated and control cells were compared at six time-points over a 48 h culture period. BMAA had a profound effect on intracellular AA levels at specific time points but in most cases, AA homeostasis was re-established in the cell. The most heavily impacted amino acid was serine which was depleted in BMAA-treated cells from 9 h onwards. Correction of serine depletion could be a factor in the observation that supplementation with L-serine protects against BMAA toxicity in vitro and in vivo. AAs that could potentially be involved in protection against BMAA-induced oxidation such as histidine, tyrosine, and phenylalanine were depleted in cells at later time points.
Collapse
Affiliation(s)
- Jake P. Violi
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| | - Lisa Pu
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| | - Sercan Pravadali-Cekic
- School of Mathematical and Physical Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia (D.P.B.)
| | - David P. Bishop
- School of Mathematical and Physical Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia (D.P.B.)
| | - Connor R. Phillips
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| | - Kenneth J. Rodgers
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW 2007, Australia; (J.P.V.); (L.P.); (C.R.P.)
| |
Collapse
|