1
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [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: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
2
|
Ding N, Song Y, Zhang Y, Yu W, Li X, Li W, Li L. Heat-shock chaperone HSPB1 mitigates poly-glycine-induced neurodegeneration via restoration of autophagic flux. Autophagy 2025; 21:1298-1315. [PMID: 39936620 DOI: 10.1080/15548627.2025.2466144] [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: 10/13/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025] Open
Abstract
The CGG repeat expansions in the 5'-UTR regions of certain genes have been implicated in various neurodegenerative and muscular disorders. However, the underlying pathogenic mechanisms are not well understood. In this study, we explore the role of the small molecular chaperone HSPB1 in counteracting neurodegeneration induced by poly-glycine (poly-G) aggregates. Employing a reporter system, we demonstrate that CGG repeat expansions within the 5'-UTR of the GIPC1 gene produce poly-G proteins, by repeat-associated non-AUG (RAN) translation. Through proximity labeling and subsequent mass spectrometry analysis, we characterize the composition of poly-G insoluble aggregates and reveal that these aggregates sequester key macroautophagy/autophagy receptors, SQSTM1/p62 and TOLLIP. This sequestration disrupts MAP1LC3/LC3 recruitment and impairs autophagosome formation, thereby compromising the autophagic pathway. Importantly, we show that HSPB1 facilitates the dissociation of these receptors from poly-G aggregates and consequently restores autophagic function. Overexpressing HSPB1 alleviates poly-G-induced neurodegeneration in mouse models. Taken together, these findings highlight a mechanistic basis for the neuroprotective effects of HSPB1 and suggest its potential as a therapeutic target in treating poly-G-associated neurodegenerative diseases.Abbreviations: AD: Alzheimer disease; AIF1/Iba1: allograft inflammatory factor 1; Baf A1: bafilomycin A1; BFP: blue fluorescent protein; CQ: chloroquine; EIF2A/eIF-2α: eukaryotic translation initiation factor 2A; FRAP: fluorescence recovery after photobleaching; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFAP: glial fibrillary acidic protein; GFP: green fluorescent protein; HSPB1: heat shock protein family B (small) member 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; NOTCH2NLC: notch 2 N-terminal like C; PD: Parkinson disease; PFA: paraformaldehyde; poly-A: poly-alanine; poly-G: poly-glycine; poly-R: poly-arginine; RAN translation: repeat-associated non-AUG translation; RBFOX3/NeuN: RNA binding fox-1 homolog 3; STED: stimulated emission depletion; TARDBP/TDP-43: TAR DNA binding protein; TG: thapsigargin; TOLLIP: toll interacting protein.
Collapse
Affiliation(s)
- Ning Ding
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yijie Song
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuhang Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wei Yu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xinnan Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Lingang Laboratory, Shanghai, China
| | - Wei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| | - Lei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| |
Collapse
|
3
|
Monteiro Neto JR, de Souza GF, Dos Santos VM, de Holanda Paranhos L, Ribeiro GD, Magalhães RSS, Queiroz DD, Eleutherio ECA. SOD1, A Crucial Protein for Neural Biochemistry: Dysfunction and Risk of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2025:10.1007/s12035-025-05067-1. [PMID: 40419749 DOI: 10.1007/s12035-025-05067-1] [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: 02/24/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025]
Abstract
Neurons are very susceptible to oxidative stress. They are the major consumers of oxygen in the brain, which is used to provide energy through oxidative phosphorylation, the major source of reactive oxygen species (ROS). In addition, compared to other tissues, neurons have lower levels of catalase and glutathione and increased susceptibility to lipid peroxidation due to the elevated levels of unsaturated fatty acids. These characteristics increasingly emphasize the antioxidant enzyme Cu/Zn superoxide dismutase 1 (SOD1) to maintain neuronal redox homeostasis. In the last decade, SOD1 gained additional roles which are also important to the metabolism of neurons. SOD1 controls the production of ROS by the electron transport chain, activates the expression of genes involved in the protection against oxidative stress, and regulates the shift from oxidative to fermentative metabolism involved in astrocyte-neuron metabolic cooperation. Furthermore, impaired interaction between the phosphatase calcineurin and SOD1 seems to result in TDP-43 hyperphosphorylation, the main proteinopathy found in amyotrophic lateral sclerosis (ALS) patients. However, this enzyme is ubiquitously expressed, mutated, and damaged forms of SOD1 cause disease in motor neurons. In this review, we discuss the pivotal functions of SOD1 in neuronal biochemistry and their implications for ALS.
Collapse
Affiliation(s)
- José Raphael Monteiro Neto
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Gabriel Freitas de Souza
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Vanessa Mattos Dos Santos
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Luan de Holanda Paranhos
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Gabriela Delaqua Ribeiro
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Rayne Stfhany Silva Magalhães
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Daniela Dias Queiroz
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil
| | - Elis Cristina Araujo Eleutherio
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Av. Athos da Silveira Ramos, 149, Rio de Janeiro, RJ, 21941-909, Brazil.
| |
Collapse
|
4
|
Schweingruber C, Nijssen J, Mechtersheimer J, Reber S, Lebœuf M, O'Brien NL, Mei I, Hedges E, Keuper M, Benitez JA, Radoi V, Jastroch M, Ruepp MD, Hedlund E. Single-cell RNA-sequencing reveals early mitochondrial dysfunction unique to motor neurons shared across FUS- and TARDBP-ALS. Nat Commun 2025; 16:4633. [PMID: 40389397 PMCID: PMC12089458 DOI: 10.1038/s41467-025-59679-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Mutations in FUS and TARDBP cause amyotrophic lateral sclerosis (ALS), but the precise mechanisms of selective motor neuron degeneration remain unresolved. To address if pathomechanisms are shared across mutations and related to either gain- or loss-of-function, we performed single-cell RNA sequencing across isogenic induced pluripotent stem cell-derived neuron types, harbouring FUS P525L, FUS R495X, TARDBP M337V mutations or FUS knockout. Transcriptional changes were far more pronounced in motor neurons than interneurons. About 20% of uniquely dysregulated motor neuron transcripts were shared across FUS mutations, half from gain-of-function. Most indicated mitochondrial impairments, with attenuated pathways shared with mutant TARDBP M337V as well as C9orf72-ALS patient motor neurons. Mitochondrial motility was impaired in ALS motor axons, even with nuclear localized FUS mutants, demonstrating shared toxic gain-of-function mechanisms across FUS- and TARDBP-ALS, uncoupled from protein mislocalization. These early mitochondrial dysfunctions unique to motor neurons may affect survival and represent therapeutic targets in ALS.
Collapse
Affiliation(s)
- Christoph Schweingruber
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16C, 106 91, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Solna v. 9, 171 77, Stockholm, Sweden
| | - Jik Nijssen
- Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Solna v. 9, 171 77, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Solna v. 9, 171 77, Stockholm, Sweden
| | - Jonas Mechtersheimer
- UK Dementia Research Institute Centre at King's College London, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Rd, SE5 9RX, London, United Kingdom
| | - Stefan Reber
- UK Dementia Research Institute Centre at King's College London, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Rd, SE5 9RX, London, United Kingdom
| | - Mélanie Lebœuf
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16C, 106 91, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Solna v. 9, 171 77, Stockholm, Sweden
| | - Niamh L O'Brien
- UK Dementia Research Institute Centre at King's College London, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Rd, SE5 9RX, London, United Kingdom
| | - Irene Mei
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16C, 106 91, Stockholm, Sweden
| | - Erin Hedges
- UK Dementia Research Institute Centre at King's College London, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Rd, SE5 9RX, London, United Kingdom
| | - Michaela Keuper
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius v. 20C, 106 91, Stockholm, Sweden
| | - Julio Aguila Benitez
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Solna v. 9, 171 77, Stockholm, Sweden
| | - Vlad Radoi
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16C, 106 91, Stockholm, Sweden
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius v. 20C, 106 91, Stockholm, Sweden
| | - Marc-David Ruepp
- UK Dementia Research Institute Centre at King's College London, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Rd, SE5 9RX, London, United Kingdom.
| | - Eva Hedlund
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16C, 106 91, Stockholm, Sweden.
- Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Solna v. 9, 171 77, Stockholm, Sweden.
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Solna v. 9, 171 77, Stockholm, Sweden.
| |
Collapse
|
5
|
Bao J, Qiu Y, Wang T, Shang L, Chu S, Jin W, Wang W, Jiang Y, Li B, Huang Y, Hou B, Sha L, You Y, Li Y, Wu M, Zou Y, Wang Y, Huo L, Qiu L, Xu Q, Feng F, Mao C, Dong L, Gao J. Predictive Accuracy of a Clinical Model for Carriage of Pathogenic/Likely Pathogenic Variants in Patients with Dementia and a Positive Family History at PUMCH. Biomedicines 2025; 13:1235. [PMID: 40427062 DOI: 10.3390/biomedicines13051235] [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: 03/30/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Background and Objectives: Identifying carriers of Pathogenic/Likely Pathogenic Variants in patients with dementia is crucial for risk stratification, particularly in individuals with a family history. This study developed and validated a clinical prediction model using whole-exome sequencing-confirmed cohorts. Methods: A total of 601 Chinese patients with dementia and a family history were enrolled at Peking Union Medical College Hospital, with 476 in a retrospective derivation cohort and 125 in a temporal validation cohort. Predictive factors included age at onset, APOE ε4 status, and family history characteristics. Model performance was assessed using discrimination and calibration metrics. Results: In the derivation cohort (median age at onset 66 years), 10.3% carried Pathogenic/Likely Pathogenic Variants. Among patients with dementia, those with age at onset < 55 years (OR 2.56, p = 0.0098), more than two affected relatives (OR 3.32, p = 0.0039), parental disease history (OR 4.72, p = 0.015), and early-onset cases in the family (OR 2.61, p = 0.0096) were positively associated with Pathogenic/Likely Pathogenic Variant carriage, whereas APOE ε4 carriage was inversely associated (OR 0.36, p = 0.0041). The model achieved an area under the curve of 0.776 (95% CI, 0.701-0.853) in the derivation cohort and 0.781 (95% CI, 0.647-0.914) in the validation cohort (median age at onset 58 years), with adequate calibration. Conclusions: This model demonstrated strong predictive performance for Pathogenic/Likely Pathogenic Variant carriage, supporting its clinical utility in guiding genetic testing. Further research is needed to refine the model.
Collapse
Affiliation(s)
- Jialu Bao
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Yuyue Qiu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Tianyi Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Li Shang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Shanshan Chu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Wei Jin
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Wenjun Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Yuhan Jiang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Bo Li
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Yixuan Huang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Longze Sha
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100730, China
| | - Yunfan You
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Yuanheng Li
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Meiqi Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Nuclear Medicine, Center for Rare Diseases Research Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yifei Wang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Li Huo
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Nuclear Medicine, Center for Rare Diseases Research Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100730, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Chenhui Mao
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Liling Dong
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| | - Jing Gao
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital Translational Medical Center, Beijing 100730, China
| |
Collapse
|
6
|
Li A, Zhang J, Ma C, Qi L, Hu Q, Li Q, Fang Y, Song J, Liu Y, Zhang Y. Endosomal protein DENND10 promotes developmental competence of neurite extension. iScience 2025; 28:112385. [PMID: 40330880 PMCID: PMC12051703 DOI: 10.1016/j.isci.2025.112385] [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: 09/10/2023] [Revised: 10/24/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
A distinguishing feature of neurons is the presence of long neurites that enable far-reaching communication. Establishing this complex morphology requires precise regulation of intracellular transport and signaling. Our study identifies DENND10, an ancient endosomal protein, as a crucial factor in shaping neuron morphology. DENND10 is a potential regulator of Rab GTPase signaling and interacts with the CCC/Retriever endosomal complex. Loss of DENND10 in a neuronal cell culture model resulted in shortened neurites. Quantitative proteomics revealed two distinct processes of neurite outgrowth: differentiation-induced biochemical changes and a pre-existing vesicular transport system modulated by DENND10. Mechanistically, both Rab27 and CCC complex subunit CCDC22 act downstream of DENND10 to support neurite extension. In primary cortical neurons, loss of DENND10 or CCDC22 led to shortened dendrites and impaired axon development. These findings provide a conceptual framework for neuronal morphogenesis during differentiation and highlight the critical role of DENND10/CCC in neurite extension.
Collapse
Affiliation(s)
- Aiqing Li
- School of Life Sciences, Suzhou Medical College of Soochow University,
Suzhou 215123, China
| | - Jie Zhang
- School of Life Sciences, Suzhou Medical College of Soochow University,
Suzhou 215123, China
| | - Chao Ma
- School of Life Sciences, Suzhou Medical College of Soochow University,
Suzhou 215123, China
| | - Lijuan Qi
- School of Life Sciences, Suzhou Medical College of Soochow University,
Suzhou 215123, China
| | - Qiuming Hu
- School of Life Sciences, Suzhou Medical College of Soochow University,
Suzhou 215123, China
| | - Qian Li
- School of Life Sciences, Suzhou Medical College of Soochow University,
Suzhou 215123, China
| | - Yufei Fang
- Wisdom Lake Academy of Pharmacy, Jiangsu Provincial Higher Education Key
Laboratory of Cell Therapy Nanoformulation (Construction), Suzhou Municipal Key
Lab of Metabolic Syndrome and Drug Research, School of Science, Xi’an
Jiaotong-Liverpool University, Suzhou 215123, China
| | - Jianrui Song
- Wisdom Lake Academy of Pharmacy, Jiangsu Provincial Higher Education Key
Laboratory of Cell Therapy Nanoformulation (Construction), Suzhou Municipal Key
Lab of Metabolic Syndrome and Drug Research, School of Science, Xi’an
Jiaotong-Liverpool University, Suzhou 215123, China
| | - Yaobo Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of
Neuroscience, Clinical Research Center of Neurological Disease, The Second
Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu
215123, China
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of
Soochow University, Suzhou 215123, China
| | - Yanling Zhang
- School of Life Sciences, Suzhou Medical College of Soochow University,
Suzhou 215123, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical
College of Soochow University, Suzhou 215123, China
| |
Collapse
|
7
|
Verma KK, Gaur PK, Gupta SL, Lata K, Kaushik R, Sharma V. Metabolomics: a new frontier in neurodegenerative disease biomarker discovery. Metabolomics 2025; 21:67. [PMID: 40374790 DOI: 10.1007/s11306-025-02267-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Neurodegenerative disorders are a group of debilitating diseases affecting the central nervous system, and are characterized by the progressive loss of neurons, leading to declines in cognitive function, movement, and overall quality of life. While the exact causes remain elusive, it's believed that a combination of genetic, environmental, and lifestyle factors contribute to their development. Metabolites, the end products of cellular processes, reflect the physiological state of an organism. By analysing these molecules, researchers can gain a deeper understanding of the underlying metabolic changes associated with neurodegenerative disorders. AIM OF REVIEW This review aims to explore the possibilities between metabolites and their association with neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), Multiple sclerosis (MS) and Huntington's disease (HD). KEY SCIENTIFIC CONCEPTS OF REVIEW Metabolomic studies could potentially illuminate altered biochemical pathways, facilitating earlier detection and treatment of these conditions. Metabolomic investigations have revealed the role of oxidative stress, alterations in glucose and fat metabolism, mitochondrial dysfunction, apoptosis, glutamate excitotoxicity and alterations in myelin composition in neurodegenerative disorders. The common metabolic biomarkers identified includes glutamate, taurine, uric acid, branched chain amino acids, acylcarnitine, creatinine, choline, with some more amino acids and lipids. Metabolomics offers valuable insights into disease mechanisms and potential therapeutic targets by identifying biochemical and metabolic alterations, but still there are several aspects to be explored for accurate mapping of metabolites with specific pathway involved in the disease.
Collapse
Affiliation(s)
- Krishan Kumar Verma
- Metro College of Health Sciences & Research, Plot No. 41, Knowledge Park-III , Greater Noida, Uttar Pradesh, India.
| | - Praveen Kumar Gaur
- Metro College of Health Sciences & Research, Plot No. 41, Knowledge Park-III , Greater Noida, Uttar Pradesh, India
| | - Sonia Lal Gupta
- Department of Neurology & Neurosurgery, Metro Hospitals, Sector-11, Noida, Uttar Pradesh, India
| | - Kanak Lata
- Metro College of Health Sciences & Research, Plot No. 41, Knowledge Park-III , Greater Noida, Uttar Pradesh, India
| | - Rahul Kaushik
- Metro College of Health Sciences & Research, Plot No. 41, Knowledge Park-III , Greater Noida, Uttar Pradesh, India
| | - Vikas Sharma
- Metro College of Health Sciences & Research, Plot No. 41, Knowledge Park-III , Greater Noida, Uttar Pradesh, India
| |
Collapse
|
8
|
Trautwig AN, Fox EJ, Dammer EB, Shantaraman A, Ping L, Duong DM, Watson CM, Wu F, Asress S, Guo Q, Levey AI, Lah JJ, Verde F, Doretti A, Ratti A, Ticozzi N, Ly CV, Miller TM, Garret MA, Berry JD, Thomas EV, Fournier CN, McEachin ZT, Seyfried NT, Glass JD. Network analysis of the cerebrospinal fluid proteome reveals shared and unique differences between sporadic and familial forms of amyotrophic lateral sclerosis. Mol Neurodegener 2025; 20:58. [PMID: 40375307 DOI: 10.1186/s13024-025-00838-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/10/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Amyotrophic Lateral Sclerosis (ALS), a neurodegenerative disease involving loss of motor neurons, typically results in death within 3-5 years of disease onset. Although roughly 10% of cases can be linked to a specific inherited mutation (e.g., C9orf72 hexanucleotide repeat expansion or SOD1 mutation), the cause(s) of most cases are unknown. Consequently, there is a critical need for biomarkers that reflect disease onset and progression across ALS subgroups. METHODS We employed tandem mass tag mass spectrometry (TMT-MS) based proteomics on cerebrospinal fluid (CSF) to identify and quantify 2105 proteins from sporadic, C9orf72, and SOD1 ALS patients, asymptomatic C9orf72 expansion carriers, and controls (N = 101). To verify trends in our Emory University cohort we used data-independent acquisition (DIA-MS) on an expanded, four center cohort. This expanded cohort of 259 individuals included 50 sporadic ALS (sALS), 43 C9orf72 ALS, 22 SOD1 ALS, 72 asymptomatic gene carriers (59 C9orf72 and 13 SOD1) and 72 age-matched controls. We identified 2330 proteins and used differential protein abundance and network analyses to determine how protein profiles vary across disease subtypes in ALS CSF. RESULTS Differential abundance and co-expression network analysis identified proteomic differences between ALS and control, as well as differentially abundant proteins between sporadic, C9orf72 and SOD1 ALS. A panel of proteins differentiated forms of ALS that are indistinguishable in a clinical setting. An additional panel differentiated asymptomatic from symptomatic C9orf72 and SOD1 mutation carriers, marking a pre-symptomatic proteomic signature of genetic forms of ALS. Leveraging this large, multicenter cohort, we validated our ALS CSF network and identified ALS-specific proteins and network modules. CONCLUSIONS This study represents a comprehensive analysis of the CSF proteome across sporadic and genetic causes of ALS that resolves differences among these ALS subgroups and also identifies proteins that distinguish symptomatic from asymptomatic gene carriers. These new data point to varying pathogenic pathways that result in an otherwise clinically indistinguishable disease.
Collapse
Affiliation(s)
- Adam N Trautwig
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Edward J Fox
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Anantharaman Shantaraman
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Lingyan Ping
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Caroline M Watson
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Fang Wu
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Seneshaw Asress
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Qi Guo
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Federico Verde
- Department of Neuroscience, IRCCS Instituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, Universitá Degli Studi Di Milano, Milan, Italy
| | - Alberto Doretti
- Department of Neuroscience, IRCCS Instituto Auxologico Italiano, Milan, Italy
| | - Antonia Ratti
- Department of Medical Biotechnology and Translational Medicine, Universitá Degli Studi Di Milano, Milan, Italy
| | - Nicola Ticozzi
- Department of Neuroscience, IRCCS Instituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, Universitá Degli Studi Di Milano, Milan, Italy
| | - Cindy V Ly
- Department of Neurology, Washington University, St Louis, MO, USA
| | - Timothy M Miller
- Department of Neurology, Washington University, St Louis, MO, USA
| | - Mark A Garret
- Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - James D Berry
- Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Eleanor V Thomas
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Zachary T McEachin
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
9
|
Hölbling BV, Gupta Y, Marchi PM, Atilano ML, Flower M, Ureña E, Goulden RA, Dobbs HK, Katona E, Mikheenko A, Giblin A, Awan AR, Fisher-Ward CL, O'Brien N, Vaizoglu D, Kempthorne L, Wilson KM, Gittings LM, Carcolé M, Ruepp MD, Mizielinska S, Partridge L, Fratta P, Tabrizi SJ, Selvaraj BT, Chandran S, Armstrong E, Whiting P, Isaacs AM. A multimodal screening platform for endogenous dipeptide repeat proteins in C9orf72 patient iPSC neurons. Cell Rep 2025; 44:115695. [PMID: 40349338 DOI: 10.1016/j.celrep.2025.115695] [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: 06/21/2023] [Revised: 11/04/2024] [Accepted: 04/23/2025] [Indexed: 05/14/2025] Open
Abstract
Repeat expansions in C9orf72 are the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia. Repeat-associated non-AUG (RAN) translation generates neurotoxic dipeptide repeat proteins (DPRs). To study endogenous DPRs, we inserted the minimal HiBiT luciferase reporter downstream of sense repeat derived DPRs polyGA or polyGP in C9orf72 patient iPSCs. We show these "DPReporter" lines sensitively and rapidly report DPR levels in lysed and live cells and optimize screening in iPSC neurons. Small-molecule screening showed the ERK1/2 activator periplocin dose dependently increases DPR levels. Consistent with this, ERK1/2 inhibition reduced DPR levels and prolonged survival in C9orf72 repeat expansion flies. CRISPR knockout screening of all human helicases revealed telomere-associated helicases modulate DPR expression, suggesting common regulation of telomeric and C9orf72 repeats. These DPReporter lines allow investigation of DPRs in their endogenous context and provide a template for studying endogenous RAN-translated proteins, at scale, in other repeat expansion disorders.
Collapse
Affiliation(s)
- Benedikt V Hölbling
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Yashica Gupta
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Paolo M Marchi
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Magda L Atilano
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, UCL, London, UK
| | - Michael Flower
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Enric Ureña
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, UCL, London, UK
| | - Rajkumar A Goulden
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, UCL, London, UK
| | - Hannah K Dobbs
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Eszter Katona
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Alla Mikheenko
- UK Dementia Research Institute at UCL, London, UK; Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Ashling Giblin
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, UCL, London, UK
| | - Ali Raza Awan
- Genomics Innovation Unit, Guy's and St Thomas' NHS Trust, London, UK; Comprehensive Cancer Centre, King's College London, London, UK
| | | | - Niamh O'Brien
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Deniz Vaizoglu
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Liam Kempthorne
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Katherine M Wilson
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Lauren M Gittings
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Mireia Carcolé
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Marc-David Ruepp
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Sarah Mizielinska
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, UCL, London, UK
| | - Pietro Fratta
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK; The Francis Crick Institute, London, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Bhuvaneish T Selvaraj
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Siddharthan Chandran
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Emma Armstrong
- Alzheimer's Research UK Drug Discovery Institute, UCL, London, UK
| | - Paul Whiting
- UK Dementia Research Institute at UCL, London, UK; Alzheimer's Research UK Drug Discovery Institute, UCL, London, UK
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK.
| |
Collapse
|
10
|
DeLano K, Sprague AC, Jandarov R, Jackson BP, Shatz R, Langevin SM, Sawyer RP. Association of plasma concentration of trace metals with frontotemporal degeneration. Front Neurol 2025; 16:1593821. [PMID: 40417119 PMCID: PMC12098025 DOI: 10.3389/fneur.2025.1593821] [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: 03/14/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025] Open
Abstract
Objective Compare the burden of heavy metals in plasma from people with frontotemporal degeneration (FTD) and healthy controls. Methods A cross-sectional study of 14 FTD cases and 28 healthy controls recruited from the University of Cincinnati. Plasma samples were sent to the Trace Element Analysis Core at Dartmouth College for assessment of 24 metals or metalloids via inductively coupled plasma mass spectrometry (ICP-MS). Unconditional logistic regression models were performed with adjustments for age (centered at the median) and sex. Results After adjusting for age and sex, there was a significant positive association of FTD with the highest tertile of Manganese (ORadjusted = 11.1, 95% CI: 1.57-132) and Chromium (ORadjusted = 9.86, 95% CI: 1.24-218). There was significant inverse associations observed between FTD and the highest tertile of Barium (ORadjusted = 0.06, 95% CI: <0.01-0.47) and Mercury (ORadjusted = 0.13, 95% CI: 0.01-0.74), with a significant inverse trend (ptrend = 0.03). Conclusion Significant associations between plasma concentration of several trace metals and FTD. The significantly elevated levels of Manganese and Chromium may suggest a role of environmental exposure in the pathogenesis of FTD. However, larger, well-designed prospective studies, along with complementary experimental work, are needed to better elucidate this relationship.
Collapse
Affiliation(s)
- Kelly DeLano
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Alex C. Sprague
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
| | - Roman Jandarov
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
| | - Brian P. Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH, United States
| | - Rhonna Shatz
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Scott M. Langevin
- Division of Hematology & Oncology, University of Vermont Larner College of Medicine, Burlington, VT, United States
- University of Vermont Cancer Center, Burlington, VT, United States
| | - Russell P. Sawyer
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
11
|
Gerring ZF, Bhalala OG, Fearnley LG, Oikari LE, White AR, Derks EM, Watson R, Yassi N, Bahlo M, Reay WR. Drug repurposing candidates for amyotrophic lateral sclerosis using common and rare genetic variants. Brain Commun 2025; 7:fcaf184. [PMID: 40395632 PMCID: PMC12089939 DOI: 10.1093/braincomms/fcaf184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/25/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative condition for which novel disease modifying therapies are urgently needed. Given the increasing bottlenecks in drug discovery pipelines, repurposing existing drugs for ALS may represent a path to expedite translation and improve disease outcomes. However, ALS is a heterogeneous disease for which the aetiology remains poorly characterized, complicating efforts to effectively repurpose drugs. We propose that the polygenic architecture of ALS genetic liability, which ranges from ultra-rare, high-impact variation to common frequency loci of small-individual effect, could be leveraged to prioritize drug repurposing candidates which are more generalizable to the ALS clinical population. Here, we utilize common and rare frequency ALS genetic risk with a novel approach to uncover therapeutic classes that may be prospective repurposing opportunities in ALS. The common variant-led analyses integrated both positional-based and functional gene-based tests on SNP-genotype data from a genome-wide association study of ALS and implicated mitogen-activated protein kinase signalling related downregulation through B-Raf inhibitors as a prospective target for repurposing. The rare variant-led approaches leveraged rare variant burden testing of exonic variation on whole genome-sequencing data from a subset of the common variant genome-wide association study cohort and prioritized B-vitamin related candidates, such as cobalamin and niacin. Clinical characterization of these putative repurposing opportunities revealed genetic support to existing biology for which related compounds are actively proceeding through ALS clinical studies. Moreover, leveraging transcriptomic data from ALS derived cell lines carrying a selection of pathogenic variants in genes that cause familial forms of ALS (C9orf72, SOD1, FUS and TARDBP) suggested that the action of B-Raf inhibitors may be of particular relevance to C9orf72 carriers, whilst the signal for B-vitamin signalling related targets was strongest in SOD1 carriers. In summary, we demonstrate the importance of considering the therapeutic actionability of both common and rare-variant mediated risk for ALS given the immense biological heterogeneity of this disorder. Future pre-clinical and clinical studies are now warranted to further characterize the tractability of these prioritized compounds.
Collapse
Affiliation(s)
- Zachary F Gerring
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Brain and Mental Health Research Program, Queensland Institute of Medical Research Berghofer, Herston, Queensland 4006, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Oneil G Bhalala
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Neurology, The Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Liam G Fearnley
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Lotta E Oikari
- Brain and Mental Health Research Program, Queensland Institute of Medical Research Berghofer, Herston, Queensland 4006, Australia
| | - Anthony R White
- Brain and Mental Health Research Program, Queensland Institute of Medical Research Berghofer, Herston, Queensland 4006, Australia
| | - Eske M Derks
- Brain and Mental Health Research Program, Queensland Institute of Medical Research Berghofer, Herston, Queensland 4006, Australia
| | - Rosie Watson
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Neurology, The Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Nawaf Yassi
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Neurology, The Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - William R Reay
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| |
Collapse
|
12
|
Weiss A, Gilbert JW, Rivera Flores IV, Belgrad J, Ferguson C, Dogan EO, Wightman N, Mocarski K, Echeverria D, Harkins AL, Summers A, Bramato B, McHugh N, Furgal R, Yamada N, Cooper D, Monopoli K, Godinho BMDC, Hassler MR, Yamada K, Greer P, Henninger N, Brown RH, Khvorova A. RNAi-mediated silencing of SOD1 profoundly extends survival and functional outcomes in ALS mice. Mol Ther 2025:S1525-0016(25)00380-6. [PMID: 40349108 DOI: 10.1016/j.ymthe.2025.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/11/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative condition, with 20% of familial and 2%-3% of sporadic cases linked to mutations in the cytosolic superoxide dismutase (SOD1) gene. Mutant SOD1 protein is toxic to motor neurons, making SOD1 gene suppression a promising approach, supported by preclinical data and the 2023 Federal Drug Administration (FDA) approval of the GapmeR ASO targeting SOD1, tofersen. Despite the approval of an ASO and the optimism it brings to the field, the pharmacodynamics and pharmacokinetics of therapeutic SOD1 modulation can be improved. Here, we developed a chemically stabilized divalent siRNA scaffold (di-siRNA) that effectively suppresses SOD1 expression in vitro and in vivo. With optimized chemical modification, it achieves remarkable CNS tissue permeation and SOD1 silencing in vivo. Administered intraventricularly, di-siRNASOD1 extended survival in SOD1-G93A ALS mice, increasing survival beyond that previously seen in these mice by ASO modalities, slowed disease progression according to the standard ALS preclinical endpoints, and attenuated ALS neuropathology. These properties offer an improved therapeutic strategy for SOD1-mediated ALS and may extend to other dominantly inherited neurological disorders.
Collapse
Affiliation(s)
- Alexandra Weiss
- Department of Neurology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - James W Gilbert
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | | | - Jillian Belgrad
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Chantal Ferguson
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Elif O Dogan
- Department of Neurology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nicholas Wightman
- Department of Neurology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Kit Mocarski
- Department of Neurology, UMass Chan Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Dimas Echeverria
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Ashley L Harkins
- Department of Neurology, UMass Chan Medical School, Worcester, MA 01605, USA; Department of Genetic & Cellular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Ashley Summers
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Brianna Bramato
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nicholas McHugh
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Raymond Furgal
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nozomi Yamada
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - David Cooper
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Kathryn Monopoli
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Bruno M D C Godinho
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Matthew R Hassler
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Ken Yamada
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Paul Greer
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nils Henninger
- Department of Neurology, UMass Chan Medical School, Worcester, MA 01605, USA; Department of Psychiatry, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Robert H Brown
- Department of Neurology, UMass Chan Medical School, Worcester, MA 01605, USA.
| | - Anastasia Khvorova
- RNA Therapeutics Institute, UMass Chan Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
13
|
Verde EM, Secco V, Ghezzi A, Mandrioli J, Carra S. Molecular Mechanisms of Protein Aggregation in ALS-FTD: Focus on TDP-43 and Cellular Protective Responses. Cells 2025; 14:680. [PMID: 40422183 DOI: 10.3390/cells14100680] [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: 03/25/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/28/2025] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) are two neurodegenerative disorders that share common genes and pathomechanisms and are referred to as the ALS-FTD spectrum. A hallmark of ALS-FTD pathology is the abnormal aggregation of proteins, including Cu/Zn superoxide dismutase (SOD1), transactive response DNA-binding protein 43 (TDP-43), fused in sarcoma/translocated in liposarcoma (FUS/TLS), and dipeptide repeat proteins resulting from C9orf72 hexanucleotide expansions. Genetic mutations linked to ALS-FTD disrupt protein stability, phase separation, and interaction networks, promoting misfolding and insolubility. This review explores the molecular mechanisms underlying protein aggregation in ALS-FTD, with a particular focus on TDP-43, as it represents the main aggregated species inside pathological inclusions and can also aggregate in its wild-type form. Moreover, this review describes the protective mechanisms activated by the cells to prevent protein aggregation, including molecular chaperones and post-translational modifications (PTMs). Understanding these regulatory pathways could offer new insights into targeted interventions aimed at mitigating cell toxicity and restoring cellular function.
Collapse
Affiliation(s)
- Enza Maria Verde
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Valentina Secco
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Andrea Ghezzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
14
|
Wang L, Ma L, Gao Z, Wang Y, Qiu J. Significance of gene therapy in neurodegenerative diseases. Front Neurosci 2025; 19:1515255. [PMID: 40406043 PMCID: PMC12095248 DOI: 10.3389/fnins.2025.1515255] [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: 10/22/2024] [Accepted: 04/10/2025] [Indexed: 05/26/2025] Open
Abstract
Gene therapy is an approach that employs vectors to deliver genetic material to target cells, aiming to correct genes with pathogenic mutations and modulate one or more genes responsible for disease progression. It holds significant value for clinical applications and offers broad market potential due to the large patient population affected by various conditions. For instance, in 2023, the Food and Drug Administration (FDA) approved 55 new drugs, including five specifically for gene therapy targeting hematologic and rare diseases. Recently, with advancements in understanding the pathogenesis and development of neurodegenerative diseases (NDDs), gene therapy has emerged as a promising avenue for treating Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and spinal muscular atrophy (SMA), particularly in personalized medicine. Notably, the FDA has approved three clinical applications for combating SMA, utilizing viral vectors delivered via intravenous and intrathecal injections. However, gene therapy for other NDDs remains in clinical trials, necessitating improvements in viral vectors, exploration of new vectors, optimization of delivery routes, and further investigation into pathogenesis to identify novel targets. This review discusses recent advancements in gene therapy for NDDs, offering insights into developing new therapeutic strategies.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Neurology, Yantai Shan Hospital, Yantai, China
| | - Lin Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao, China
| | - Zihan Gao
- Department of Internal Medicine of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Wang
- Department of Neurology, Yantai Shan Hospital, Yantai, China
| | - Jiaoxue Qiu
- Department of Neurology, Yantai Shan Hospital, Yantai, China
| |
Collapse
|
15
|
Wu X, Yang Z, Zou J, Gao H, Shao Z, Li C, Lei P. Protein kinases in neurodegenerative diseases: current understandings and implications for drug discovery. Signal Transduct Target Ther 2025; 10:146. [PMID: 40328798 PMCID: PMC12056177 DOI: 10.1038/s41392-025-02179-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases (e.g., Alzheimer's, Parkinson's, Huntington's disease, and Amyotrophic Lateral Sclerosis) are major health threats for the aging population and their prevalences continue to rise with the increasing of life expectancy. Although progress has been made, there is still a lack of effective cures to date, and an in-depth understanding of the molecular and cellular mechanisms of these neurodegenerative diseases is imperative for drug development. Protein phosphorylation, regulated by protein kinases and protein phosphatases, participates in most cellular events, whereas aberrant phosphorylation manifests as a main cause of diseases. As evidenced by pharmacological and pathological studies, protein kinases are proven to be promising therapeutic targets for various diseases, such as cancers, central nervous system disorders, and cardiovascular diseases. The mechanisms of protein phosphatases in pathophysiology have been extensively reviewed, but a systematic summary of the role of protein kinases in the nervous system is lacking. Here, we focus on the involvement of protein kinases in neurodegenerative diseases, by summarizing the current knowledge on the major kinases and related regulatory signal transduction pathways implicated in diseases. We further discuss the role and complexity of kinase-kinase networks in the pathogenesis of neurodegenerative diseases, illustrate the advances of clinical applications of protein kinase inhibitors or novel kinase-targeted therapeutic strategies (such as antisense oligonucleotides and gene therapy) for effective prevention and early intervention.
Collapse
Affiliation(s)
- Xiaolei Wu
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhangzhong Yang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinjun Zou
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
16
|
Maiocchi A, Pedrini M, Ferrari V, Assunçao Carreira AS, D'Amore VM, Santoro F, Di Porzio A, Bosetti M, Cristofani R, Silvani A, Brancaccio D, Marinelli L, Di Leva FS, Provenzani A, Poletti A, Seneci P. Design, synthesis and characterization of aryl bis-guanyl hydrazones as RNA binders of C9orf72 G 4C 2 extended repeats. Eur J Med Chem 2025; 293:117736. [PMID: 40349639 DOI: 10.1016/j.ejmech.2025.117736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Expanded G4C2 repeats derived from mutations of the C9orf72 gene are causative factors in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients, leading to multiple pathological events. Bis thiophene para dinicotinimidamide 2a was reported to preferentially stabilize G-quadruplex G4C2 RNA structures at sub-micromolar concentrations. We replaced its amidine groups with BBB-compliant guanyl hydrazones, and carried out scaffold variations to improve water solubility. An eight-membered array was built around bis-thiophene- (4b-6a), bis-oxazole- (7b), diphenylurea diamide- (8b) and phenyldioxy ditriazolephenyl scaffolds (9a,b). Biological profiling of the array identified 4b as a promising, drug-like hit, active in cellular assays on ALS patient-derived cells.
Collapse
Affiliation(s)
- Alice Maiocchi
- Chemistry Department, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Martina Pedrini
- Chemistry Department, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DisFeB) "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Agata Sofia Assunçao Carreira
- Laboratory of Genomic Screening, Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, Povo, 38123, (TN), Italy
| | - Vincenzo Maria D'Amore
- Department of Pharmacy, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy
| | - Federica Santoro
- Department of Pharmacy, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy
| | - Anna Di Porzio
- Department of Pharmacy, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy
| | - Maddalena Bosetti
- Laboratory of Genomic Screening, Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, Povo, 38123, (TN), Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DisFeB) "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Alessandra Silvani
- Chemistry Department, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Diego Brancaccio
- Department of Pharmacy, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy
| | - Luciana Marinelli
- Department of Pharmacy, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy
| | - Francesco Saverio Di Leva
- Department of Pharmacy, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy.
| | - Alessandro Provenzani
- Laboratory of Genomic Screening, Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, Povo, 38123, (TN), Italy.
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DisFeB) "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| | - Pierfausto Seneci
- Chemistry Department, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy.
| |
Collapse
|
17
|
Kubinski S, Claus L, Schüning T, Zeug A, Kalmbach N, Staege S, Gschwendtberger T, Petri S, Wegner F, Claus P, Hensel N. Aggregates associated with amyotrophic lateral sclerosis sequester the actin-binding protein profilin 2. Hum Mol Genet 2025; 34:882-893. [PMID: 40063831 DOI: 10.1093/hmg/ddaf020] [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] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 05/08/2025] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease characterized by the degeneration of upper and lower motoneurons. The four most frequently mutated genes causing familial ALS (fALS) are C9orf72, FUS, SOD1, and TARDBP. Some of the related wild-type proteins comprise intrinsically disordered regions (IDRs) which favor their assembly in liquid droplets-the biophysical mechanism behind the formation of physiological granules such as stress granules (SGs). SGs assemble and dissolve dependent on the cellular condition. However, it has been suggested that transition from reversible SGs to irreversible aggregates contributes to the toxic properties of ALS-related mutated proteins. Sequestration of additional proteins within these aggregates may then result in downstream toxicity. While the exact downstream mechanisms remain elusive, rare ALS-causing mutations in the actin binding protein profilin 1 suggest an involvement of the actin cytoskeleton. Here, we hypothesize that profilin isoforms become sequestered in aggregates of ALS-associated proteins which induce subsequent dysregulation of the actin cytoskeleton. Interestingly, localization of neuronal profilin 2 in SGs was more pronounced compared with the ubiquitously expressed profilin 1. Accordingly, FUS and C9orf72 aggregates prominently sequestered profilin 2 but not profilin 1. Moreover, we observed a distinct sequestration of profilin 2 and G-actin to C9orf72 aggregates in different cellular models. On the functional level, we identified dysregulated actin dynamics in cells with profilin 2-sequestering aggregates. In summary, our results suggest a more common involvement of profilins in ALS pathomechanisms than indicated from the rarely occurring profilin mutations.
Collapse
Affiliation(s)
- Sabrina Kubinski
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Center for Systems Neuroscience (ZSN), Buenteweg 2, 30559 Hannover, Germany
| | - Luisa Claus
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Tobias Schüning
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Center for Systems Neuroscience (ZSN), Buenteweg 2, 30559 Hannover, Germany
| | - Andre Zeug
- Hannover Medical School, Cellular Neurophysiology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Norman Kalmbach
- Hannover Medical School, Department of Neurology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Selma Staege
- Center for Systems Neuroscience (ZSN), Buenteweg 2, 30559 Hannover, Germany
- Hannover Medical School, Department of Neurology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Thomas Gschwendtberger
- Center for Systems Neuroscience (ZSN), Buenteweg 2, 30559 Hannover, Germany
- Hannover Medical School, Department of Neurology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Susanne Petri
- Center for Systems Neuroscience (ZSN), Buenteweg 2, 30559 Hannover, Germany
- Hannover Medical School, Department of Neurology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Florian Wegner
- Center for Systems Neuroscience (ZSN), Buenteweg 2, 30559 Hannover, Germany
- Hannover Medical School, Department of Neurology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Peter Claus
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Center for Systems Neuroscience (ZSN), Buenteweg 2, 30559 Hannover, Germany
- Hannover Medical School, Laboratory of Molecular Neurosciences, Department of Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Niko Hensel
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Faculty of Medicine, Große Steinstraße 52, 06108 Halle (Saale), Germany
| |
Collapse
|
18
|
Serpente M, Delvecchio G, Fenoglio C, Di Consoli L, Giudici G, Borracci V, Rotondo E, Arcaro M, Sacchi L, Pintus M, Ghezzi L, Ferro A, Prunas C, Callari A, Scola E, Triulzi FM, Arighi A, Brambilla P, Galimberti D. Differential miRNA expression in neural-enriched extracellular vesicles as potential biomarker for frontotemporal dementia and bipolar disorder. Neurobiol Dis 2025; 208:106867. [PMID: 40064379 DOI: 10.1016/j.nbd.2025.106867] [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: 01/14/2025] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025] Open
Abstract
Behavioral variant of Frontotemporal Dementia (bvFTD) and Bipolar Disorder (BD) share overlapping symptoms, complicating diagnosis. BvFTD, especially linked to C9orf72 expansions, often mimics BD, highlighting the need for reliable biomarkers. This study aimed to differentiate bvFTD from BD using miRNA profiles in neural-enriched extracellular vesicles (NEVs). A cohort of 100 subjects was analyzed: 40 bvFTD (20 sporadic, 20 C9orf72 carriers), 40 BD, and 20 healthy controls. NEVs were isolated from plasma and profiled using real-time PCR. Among 754 miRNAs, 11 were significantly deregulated in bvFTD and BD. MiR-152-5p was downregulated in sporadic bvFTD, while let-7b, let-7e, miR-18b, and miR-142-5p were altered in C9orf72 carriers. BD patients showed distinct patterns in miR-331-5p, miR-335, and miR-345 compared to bvFTD. Bioinformatics analyses revealed that let-7e, let-7b, miR-18b, and miR-142-5p share common long non-coding RNA (lncRNA) targets, including XIST, NEAT1, and OIP5-AS1, suggesting their involvement in molecular networks relevant to C9orf72-related bvFTD. These miRNA signatures can differentiate bvFTD from BD, especially in C9orf72-related cases, and offer insights into disease pathways. Further research is needed to validate these findings and explore their clinical application.
Collapse
Affiliation(s)
- Maria Serpente
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy.
| | - Giuseppe Delvecchio
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Lorena Di Consoli
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Giulia Giudici
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Vittoria Borracci
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Emanuela Rotondo
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Marina Arcaro
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sacchi
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Manuela Pintus
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Laura Ghezzi
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Adele Ferro
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Cecilia Prunas
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Antonio Callari
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Elisa Scola
- Neuroradiology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio M Triulzi
- Neuroradiology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Dept of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Andrea Arighi
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Brambilla
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy; Dept of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| |
Collapse
|
19
|
Fu Y, Zhang J, Qin R, Ren Y, Zhou T, Han B, Liu B. Activating autophagy to eliminate toxic protein aggregates with small molecules in neurodegenerative diseases. Pharmacol Rev 2025; 77:100053. [PMID: 40187044 DOI: 10.1016/j.pharmr.2025.100053] [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: 12/31/2023] [Accepted: 12/05/2024] [Indexed: 04/07/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are well known to pose formidable challenges for their treatment due to their intricate pathogenesis and substantial variability among patients, including differences in environmental exposures and genetic predispositions. One of the defining characteristics of NDs is widely reported to be the buildup of misfolded proteins. For example, Alzheimer disease is marked by amyloid beta and hyperphosphorylated Tau aggregates, whereas Parkinson disease exhibits α-synuclein aggregates. Amyotrophic lateral sclerosis and frontotemporal dementia exhibit TAR DNA-binding protein 43, superoxide dismutase 1, and fused-in sarcoma protein aggregates, and Huntington disease involves mutant huntingtin and polyglutamine aggregates. These misfolded proteins are the key biomarkers of NDs and also serve as potential therapeutic targets, as they can be addressed through autophagy, a process that removes excess cellular inclusions to maintain homeostasis. Various forms of autophagy, including macroautophagy, chaperone-mediated autophagy, and microautophagy, hold a promise in eliminating toxic proteins implicated in NDs. In this review, we focus on elucidating the regulatory connections between autophagy and toxic proteins in NDs, summarizing the cause of the aggregates, exploring their impact on autophagy mechanisms, and discussing how autophagy can regulate toxic protein aggregation. Moreover, we underscore the activation of autophagy as a potential therapeutic strategy across different NDs and small molecules capable of activating autophagy pathways, such as rapamycin targeting the mTOR pathway to clear α-synuclein and Sertraline targeting the AMPK/mTOR/RPS6KB1 pathway to clear Tau, to further illustrate their potential in NDs' therapeutic intervention. Together, these findings would provide new insights into current research trends and propose small-molecule drugs targeting autophagy as promising potential strategies for the future ND therapies. SIGNIFICANCE STATEMENT: This review provides an in-depth overview of the potential of activating autophagy to eliminate toxic protein aggregates in the treatment of neurodegenerative diseases. It also elucidates the fascinating interrelationships between toxic proteins and the process of autophagy of "chasing and escaping" phenomenon. Moreover, the review further discusses the progress utilizing small molecules to activate autophagy to improve the efficacy of therapies for neurodegenerative diseases by removing toxic protein aggregates.
Collapse
Affiliation(s)
- Yuqi Fu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueting Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Brain Science, Faculty of Medicine, Imperial College, London, UK
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
Chong ZZ, Souayah N. Targeting Gene C9orf72 Pathogenesis for Amyotrophic Lateral Sclerosis. Int J Mol Sci 2025; 26:4276. [PMID: 40362512 PMCID: PMC12072292 DOI: 10.3390/ijms26094276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult neurodegenerative disorder. Since no cure has been found, finding effective therapeutic targets for ALS remains a major challenge. Gene C9orf72 mutations with the formation of hexanucleotide repeat (GGGGCC) expansion (HRE) have been considered the most common genetic pathogenesis of ALS. The literature review indicates that the C9orf72 HRE causes both the gain-of-function toxicity and loss of function of C9ORF72. The formation of RNA foci and dipeptide repeats (DPRs) resulting from HRE is responsible for toxic function gain. The RNA foci can interfere with RNA processing, while DPRs directly bind to and sequester associated proteins to disrupt processes of rRNA synthesis, mRNA translation, autophagy, and nucleocytoplasmic transport. The mutations of C9orf72 and HRE result in the loss of functional C9ORF72. Under physiological conditions, C9ORF72 binds to Smith-Magenis chromosome region 8 and WD repeat-containing protein and forms a protein complex. Loss of C9ORF72 leads to autophagic impairment, increased oxidative stress, nucleocytoplasmic transport impairment, and inflammatory response. The attempted treatments for ALS have been tried by targeting C9orf72 HRE; however, the outcomes are far from satisfactory yet. More studies should be performed on pharmacological and molecular modulators against C9orf72 HRE to evaluate their efficacy by targeting HRE.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Department of Neurology, New Jersey Medical School, Rutgers University, 185 S Orange, Newark, NJ 07103, USA
| | - Nizar Souayah
- Department of Neurology, New Jersey Medical School, Rutgers University, 90 Bergen Street DOC 8100, Newark, NJ 07101, USA
| |
Collapse
|
21
|
Grima N, Smith AN, Shepherd CE, Henden L, Zaw T, Carroll L, Rowe DB, Kiernan MC, Blair IP, Williams KL. Multi-region brain transcriptomic analysis of amyotrophic lateral sclerosis reveals widespread RNA alterations and substantial cerebellum involvement. Mol Neurodegener 2025; 20:40. [PMID: 40275359 PMCID: PMC12023386 DOI: 10.1186/s13024-025-00820-5] [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] [Received: 08/14/2024] [Accepted: 02/26/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that primarily affects the motor neurons, causing progressive muscle weakness and paralysis. While research has focused on understanding pathological mechanisms in the motor cortex and spinal cord, there is growing evidence that extra-motor brain regions may also play a role in the pathogenesis or progression of ALS. METHODS We generated 165 sample-matched post-mortem brain transcriptomes from 22 sporadic ALS patients with pTDP-43 pathological staging and 11 non-neurological controls. For each individual, five brain regions underwent mRNA sequencing: motor cortex (pTDP-43 inclusions always present), prefrontal cortex and hippocampus (pTDP-43 inclusions sometimes present), and occipital cortex and cerebellum (pTDP-43 inclusions rarely present). We examined gene expression, cell-type composition, transcript usage (% contribution of a transcript to total gene expression) and alternative splicing, comparing ALS-specific changes between brain regions. We also considered whether post-mortem pTDP-43 pathological stage classification defined ALS subgroups with distinct gene expression profiles. RESULTS Significant gene expression changes were observed in ALS cases for all five brain regions, with the cerebellum demonstrating the largest number of total (> 3,000) and unique (60%) differentially expressed genes. Pathway enrichment and predicted activity were largely concordant across brain regions, suggesting that ALS-linked mechanisms, including inflammation, mitochondrial dysfunction and oxidative stress, are also dysregulated in non-motor brain regions. Switches in transcript usage were identified for a small set of genes including increased usage of a POLDIP3 transcript, associated with TDP-43 loss-of-function, in the cerebellum and a XBP1 transcript, indicative of unfolded protein response activity, in the motor cortex. Extensive variation in RNA splicing was identified in the ALS brain, with 26-41% of alternatively spliced genes unique to a given brain region. This included detection of TDP-43-associated cryptic splicing events such as the STMN2 cryptic exon which was shown to have a pTDP-43 pathology-specific expression pattern. Finally, ALS patients with stage 4 pTDP-43 pathology demonstrated distinct gene and protein expression changes in the cerebellum. CONCLUSIONS Together our findings highlighted widespread transcriptome alterations in ALS post-mortem brain and showed that, despite the absence of pTDP-43 pathology in the cerebellum, extensive and pTDP-43 pathological stage-specific RNA changes are evident in this brain region.
Collapse
Affiliation(s)
- Natalie Grima
- Macquarie University Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Andrew N Smith
- Macquarie University Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Lyndal Henden
- Macquarie University Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Thiri Zaw
- Australian Proteome Research Facility, Macquarie University, Sydney, NSW, 2109, Australia
| | - Luke Carroll
- Australian Proteome Research Facility, Macquarie University, Sydney, NSW, 2109, Australia
| | - Dominic B Rowe
- Macquarie University Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Matthew C Kiernan
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, 2050, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, 2050, Australia
| | - Ian P Blair
- Macquarie University Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Kelly L Williams
- Macquarie University Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
22
|
Shiryaeva O, Tolochko C, Alekseeva T, Dyachuk V. Targets and Gene Therapy of ALS (Part 1). Int J Mol Sci 2025; 26:4063. [PMID: 40362304 PMCID: PMC12071412 DOI: 10.3390/ijms26094063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the selective death of motor neurons, which causes muscle atrophy. Genetic forms of ALS are recorded only in 10% of cases. However, over the past decade, studies in genetics have substantially contributed to our understanding of the molecular mechanisms underlying ALS. The identification of key mutations such as SOD1, C9orf72, FUS, and TARDBP has led to the development of targeted therapy that is gradually being introduced into clinical trials, opening up a broad range of opportunities for correcting these mutations. In this review, we aimed to present an extensive overview of the currently known mechanisms of motor neuron degeneration associated with mutations in these genes and also the gene therapy methods for inhibiting the expression of their mutant proteins. Among these, antisense oligonucleotides, RNA interference (siRNA and miRNA), and gene-editing (CRISPR/Cas9) methods are of particular interest. Each has shown its efficacy in animal models when targeting mutant genes, whereas some of them have proven to be efficient in human clinical trials.
Collapse
Affiliation(s)
| | | | | | - Vyacheslav Dyachuk
- Almazov Federal Medical Research Centre, 197341 Saint Petersburg, Russia; (O.S.); (C.T.); (T.A.)
| |
Collapse
|
23
|
Pottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Tempini MLG, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene CD, Jin LW, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, et alPottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Tempini MLG, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene CD, Jin LW, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, Van Deerlin V, Castellani R, Mesulam MM, Tartaglia MC, Finger EC, Troakes C, Al-Sarraj S, Dalgard CL, Miller BL, Seelaar H, Graff-Radford NR, Boeve BF, Mackenzie IR, van Swieten JC, Seeley WW, Sleegers K, Dickson DW, Biernacka JM, Rademakers R. Deciphering distinct genetic risk factors for FTLD-TDP pathological subtypes via whole-genome sequencing. Nat Commun 2025; 16:3914. [PMID: 40280976 PMCID: PMC12032271 DOI: 10.1038/s41467-025-59216-0] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Frontotemporal lobar degeneration with neuronal inclusions of the TAR DNA-binding protein 43 (FTLD-TDP) is a fatal neurodegenerative disorder with only a limited number of risk loci identified. We report our comprehensive genome-wide association study as part of the International FTLD-TDP Whole-Genome Sequencing Consortium, including 985 patients and 3,153 controls compiled from 26 institutions/brain banks in North America, Europe and Australia, and meta-analysis with the Dementia-seq cohort. We confirm UNC13A as the strongest overall FTLD-TDP risk factor and identify TNIP1 as a novel FTLD-TDP risk factor. In subgroup analyzes, we further identify genome-wide significant loci specific to each of the three main FTLD-TDP pathological subtypes (A, B and C), as well as enrichment of risk loci in distinct tissues, brain regions, and neuronal subtypes, suggesting distinct disease aetiologies in each of the subtypes. Rare variant analysis confirmed TBK1 and identified C3AR1, SMG8, VIPR1, RBPJL, L3MBTL1 and ANO9, as novel subtype-specific FTLD-TDP risk genes, further highlighting the role of innate and adaptive immunity and notch signaling pathway in FTLD-TDP, with potential diagnostic and novel therapeutic implications.
Collapse
Affiliation(s)
- Cyril Pottier
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA.
| | - Fahri Küçükali
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Matt Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Gregory D Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Cristina T Vicente
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Wouter De Coster
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Sarah Wynants
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Pieter Van de Walle
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Júlia Faura
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Stephen J Haggarty
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Merel O Mol
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ging-Yuek R Hsiung
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Caroline Graff
- Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital, Solna, Sweden
| | - Linn Öijerstedt
- Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital, Solna, Sweden
| | - Manuela Neumann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, University of Tübingen, Tübingen, Germany
| | - Yan Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Saurabh Baheti
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Kevin F Bieniek
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Leah Forsberg
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Hilary Heuer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Argentina Lario Lago
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Ethan G Geier
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Jennifer S Yokoyama
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Alexis P Oddi
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Margaret Flanagan
- University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - John R Hodges
- Central Clinical School and Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - John B Kwok
- University of Sydney, Sydney, NSW, Australia
- NeuRA, University of New South Wales, Randwick, NSW, Australia
| | | | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Carlo Wilke
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Chiadi Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | | | - Bret M Evers
- Division of Neuropathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
| | - David G Munoz
- St. Michael's Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Julia Keith
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Lorne Zinman
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ekaterina Rogaeva
- Krembil Discovery Tower, Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
| | - EunRan Suh
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
- kbo-Inn-Salzach-Klinikum, Clinical Center for Psychiatry, Psychotherapy, Psychosomatic Medicine, Geriatrics and Neurology, Wasserburg/Inn, Germany
| | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | | | - Laura L Donker Kaat
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Edward D Huey
- Department of Psychiatry and Human Behavior, Brown Alpert Medical School, Brown University, Providence, RI, USA
| | | | - Simon Mead
- MRC Prion Unit at University College London, Institute of Prion Diseases, London, UK
| | - Andrew King
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, London, UK
| | - Sigrun Roeber
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Alissa L Nana
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Eliana Marisa Ramos
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lea T Grinberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Maria Luisa Gorno Tempini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Howard J Rosen
- Department of Pathology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Olivier Piguet
- School of Psychology and Brain and Mind Centre, University of Sydney, Sydney, SWA, Australia
| | - Murray Grossman
- Department of Neurology, Penn Frontotemporal Degeneration Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - C Dirk Keene
- University of Washington BioRepository and Integrated Neuropathology (BRaIN) lab, Harborview Medical Center, Seattle, WA, USA
| | - Lee-Way Jin
- M.I.N.D. Institute Laboratory, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Johannes Prudlo
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Robert A Rissman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas Arzberger
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Adam L Boxer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Lawrence S Honig
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Jean P Vonsattel
- Department of Pathology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles L White
- Division of Neuropathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marla Gearing
- Department of Pathology and Laboratory Medicine and Department of Neurology, Emory University, Atlanta, GA, USA
| | - Jonathan Glass
- Department of Pathology and Laboratory Medicine and Department of Neurology, Emory University, Atlanta, GA, USA
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, University College London, Queen Square Institute of Neurology, London, UK
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Vivianna Van Deerlin
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rudolph Castellani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marsel M Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Maria C Tartaglia
- Krembil Discovery Tower, Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
| | - Elizabeth C Finger
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Safa Al-Sarraj
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Bruce L Miller
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Ian Ra Mackenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - William W Seeley
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Kristel Sleegers
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | | | - Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
| |
Collapse
|
24
|
Hile SE, Weissensteiner MH, Pytko KG, Dahl J, Kejnovsky E, Kejnovská I, Hedglin M, Georgakopoulos-Soares I, Makova K, Eckert KA. Replicative DNA polymerase epsilon and delta holoenzymes show wide-ranging inhibition at G-quadruplexes in the human genome. Nucleic Acids Res 2025; 53:gkaf352. [PMID: 40298112 PMCID: PMC12038398 DOI: 10.1093/nar/gkaf352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
G-quadruplexes (G4s) are functional elements of the human genome, some of which inhibit DNA replication. We investigated replication of G4s within highly abundant microsatellite (GGGA, GGGT) and transposable element (L1 and SVA) sequences. We found that genome-wide, numerous motifs are located preferentially on the replication leading strand and the transcribed strand templates. We directly tested replicative polymerase ϵ and δ holoenzyme inhibition at these G4s, compared to low abundant motifs. For all G4s, DNA synthesis inhibition was higher on the G-rich than C-rich strand or control sequence. No single G4 was an absolute block for either holoenzyme; however, the inhibitory potential varied over an order of magnitude. Biophysical analyses showed the motifs form varying topologies, but replicative polymerase inhibition did not correlate with a specific G4 structure. Addition of the G4 stabilizer pyridostatin severely inhibited forward polymerase synthesis specifically on the G-rich strand, enhancing G/C strand asynchrony. Our results reveal that replicative polymerase inhibition at every G4 examined is distinct, causing complementary strand synthesis to become asynchronous, which could contribute to slowed fork elongation. Altogether, we provide critical information regarding how replicative eukaryotic holoenzymes navigate synthesis through G4s naturally occurring thousands of times in functional regions of the human genome.
Collapse
Affiliation(s)
- Suzanne E Hile
- Department of Pathology, The Jake Gittlen Laboratories for Cancer Research, Penn State University College of Medicine, Hershey, PA 17033, United States
| | - Matthias H Weissensteiner
- Department of Biology, Penn State University Eberly College of Science, University Park, PA 16802, United States
| | - Kara G Pytko
- Department of Chemistry, Penn State University Eberly College of Science, University Park, PA 16802, United States
| | - Joseph Dahl
- National Institute of Environmental Health Sciences, Z01 ES065070, Durham, NC 27709, United States
| | - Eduard Kejnovsky
- Department of Plant Developmental Genetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, 61265, Czech Republic
| | - Iva Kejnovská
- Department of Biophysics of Nucleic Acids, Institute of Biophysics of the Czech Academy of Sciences, Brno, 61265, Czech Republic
| | - Mark Hedglin
- Department of Chemistry, Penn State University Eberly College of Science, University Park, PA 16802, United States
| | - Ilias Georgakopoulos-Soares
- Department of Biochemistry and Molecular Biology, Penn State University College of Medicine, Hershey, PA, 17033, United States
| | - Kateryna D Makova
- Department of Biology, Penn State University Eberly College of Science, University Park, PA 16802, United States
| | - Kristin A Eckert
- Department of Pathology, The Jake Gittlen Laboratories for Cancer Research, Penn State University College of Medicine, Hershey, PA 17033, United States
| |
Collapse
|
25
|
Mirza SS, Pasternak M, Paterson AD, Rogaeva E, Tartaglia MC, Mitchell SB, Black SE, Freedman M, Tang-Wai D, Bouzigues A, Russell LL, Foster PH, Ferry-Bolder E, Bocchetta M, Cash DM, Zetterberg H, Sogorb-Esteve A, van Swieten J, Jiskoot LC, Seelaar H, Sanchez-Valle R, Laforce R, Graff C, Galimberti D, Vandenberghe R, de Mendonça A, Tiraboschi P, Santana I, Gerhard A, Levin J, Sorbi S, Otto M, Pasquier F, Ducharme S, Butler C, Le Ber I, Finger E, Rowe JB, Synofzik M, Moreno F, Borroni B, Rohrer JD, Masellis M. Disease-modifying effects of TMEM106B in genetic frontotemporal dementia: a longitudinal GENFI study. Brain 2025:awaf019. [PMID: 40260680 DOI: 10.1093/brain/awaf019] [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: 07/04/2024] [Revised: 10/31/2024] [Accepted: 12/14/2024] [Indexed: 04/24/2025] Open
Abstract
Common variants within TMEM106B are associated with risk for frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP). The G allele of the top single nucleotide polymorphism, rs1990622, confers protection against FTLD-TDP, including genetic cases due to GRN mutations or C9orf72 hexanucleotide repeat expansions. However, the effects of interaction between TMEM106B-rs1990622 and frontotemporal dementia (FTD) mutations on disease endophenotypes in genetic FTD are unknown. This longitudinal cohort study was embedded within the GENetic Frontotemporal dementia Initiative (GENFI). We included 518 participants from 222 families [209 non-carriers; 222 presymptomatic carriers (C9orf72 = 79; GRN = 101, MAPT = 42); 87 symptomatic carriers (C9orf72 = 45; GRN = 29; MAPT = 13)] followed for up to 7 years. Using linear mixed-effects models, we examined the effects of a triple interaction between TMEM106B-rs1990622G allele dosage (additive model: 0, 1 or 2 alleles) and autosomal dominant FTD mutations with clinical status, and time from baseline on (i) grey matter volume using a voxel-based analysis; (ii) serum neurofilament light chain (NfL) levels; and (iii) cognitive and behavioural measures. Mean age of participants was 47.9 ± 13.8 years, 58.1% were female and 61% had at least one G allele. C9orf72: rs1990622G allele dosage was associated with less atrophy within the right occipital region in presymptomatic carriers at baseline, and reduced atrophy rate within putamen and caudate nucleus, right frontotemporal regions, left cingulate and bilateral insular cortices in symptomatic carriers over time; lower NfL levels in presymptomatic carriers at baseline; better executive functions and language abilities in presymptomatic carriers; and maintained overall cognitive functions and behaviour in symptomatic carriers over time. GRN: rs1990622G allele dosage was associated with reduced grey matter atrophy rate within the right temporal and occipital regions in presymptomatic carriers, and within the right frontal cortex and insula over time in symptomatic carriers; lower serum NfL levels over time in presymptomatic carriers and lower NfL levels at both baseline and over time in symptomatic carriers; and better global cognitive performance at baseline and higher attention/processing speed scores over time in symptomatic carriers. MAPT: rs1990622G allele dosage was associated with reduced grey matter atrophy rate within the right inferior frontal gyrus in symptomatic carriers, but no effects on serum NfL or cognitive/behavioural measures. TMEM106B-rs1990622G allele dosage showed protective effects on multiple endophenotypes predominantly in GRN and C9orf72 groups. Therefore, TMEM106B genotype should be assessed in clinical trials, particularly of GRN- and C9orf72-related genetic FTD, due to its modifying effects on biomarker, imaging, cognitive and clinical outcomes.
Collapse
Affiliation(s)
- Saira S Mirza
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON M4N 3M5, Canada
- L. C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Maurice Pasternak
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Andrew D Paterson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 1E8, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Maria C Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
- Memory Clinic, University Health Network, Toronto, ON M5T 2S8, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Sara B Mitchell
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Sandra E Black
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON M4N 3M5, Canada
- L. C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON M6A 2E1, Canada
| | - Morris Freedman
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON M6A 2E1, Canada
| | - David Tang-Wai
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON M6A 2E1, Canada
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Phoebe H Foster
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Eve Ferry-Bolder
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Henrik Zetterberg
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg 405 30, Sweden
- Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg 413 45, Sweden
- DRI Fluid Biomarker Laboratory, Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 999 077, China
| | - Aitana Sogorb-Esteve
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
- UK Dementia Research Institute at University College London, London NW1 3BT, UK
| | - John van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam 3015 GD, The Netherlands
| | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Center, Rotterdam 3015 GD, The Netherlands
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Center, Rotterdam 3015 GD, The Netherlands
| | - Raquel Sanchez-Valle
- Alzheimer's disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona 08036, Spain
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Quebec, QC G1V 4G2, Canada
| | - Caroline Graff
- Department of Neurobiology, Care Sciences and Society; Center for Alzheimer Research, Division of Neurogeriatrics, Bioclinicum, Karolinska Institutet, Solna 171 65, Sweden
- Unit for Hereditary Dementias, Theme Inflammation and Aging, Karolinska University Hospital, Stockholm SE-171 76, Sweden
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan 20122, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Centro Dino Ferrari, Milan 20122, Italy
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven 3001, Belgium
- Neurology Service, University Hospitals Leuven, Leuven 3000, Belgium
| | | | - Pietro Tiraboschi
- Dipartimento Diagnostica e Tecnologia - Neurologia V, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano 20133, Italy
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra 3004-531, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-531, Portugal
| | - Alexander Gerhard
- Division of Psychology Communication and Human Neuroscience, Wolfson Molecular Imaging Centre, University of Manchester, Manchester M20 3LJ, UK
- Department of Nuclear Medicine, Center for Translational Neuro- and Behavioural Sciences, University Medicine Essen, Essen 45147, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians Universität München, Munich 80802, Germany
- Clinical Study Unit, German Center for Neurodegenerative Diseases (DZNE), Munich 81377, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich 81377, Germany
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, Florence 50139, Italy
- Department of Neurology, IRCCS Fondazione Don Carlo Gnocchi, Florence 50139, Italy
| | - Markus Otto
- Department of Neurology, University of Ulm, ULM 45 89081, Germany
| | - Florence Pasquier
- Division of Neurology, Faculty of Medicine, Univ Lille, Lille 59000, France
- Lille Neurosciences and Cognition, Inserm 1172, Lille 59000, France
| | - Simon Ducharme
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H4H 1R3, Canada
| | - Chris Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford OX3 7JX, UK
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière, Paris 75013, France
- Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière, Paris 75013, France
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON N6A 3K7, Canada
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, CB2 3EB, UK
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen 72076, Germany
- Faculty of Medicine, Center for Neurodegenerative Diseases (DZNE), Tübingen 72076, Germany
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Hospital Universitario Donostia, Donostia, Gipuzkoa 20014, Spain
- Center for Biomedical Research in Neurodegenerative Disease (CIBERNED), Carlos III Health Institute, Madrid 28029, Spain
- Biogipuzkoa Health Research Institute, Neurosciences Area, Group of Neurodegenerative Diseases, San Sebastian 20014, Spain
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 15-25121, Italy
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Mario Masellis
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON M4N 3M5, Canada
- L. C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
26
|
Li HX, He YM, Fei J, Guo M, Zeng C, Yan PJ, Xu Y, Qin G, Teng FY. The G-quadruplex ligand CX-5461: an innovative candidate for disease treatment. J Transl Med 2025; 23:457. [PMID: 40251554 PMCID: PMC12007140 DOI: 10.1186/s12967-025-06473-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
The ribosomal DNA (rDNA) plays a vital role in regulating protein synthesis by ribosome biogenesis, essential for maintaining cellular growth, metabolism, and more. Cancer cells show a high dependence on ribosome biogenesis and exhibit elevated rDNA transcriptional activity. CX-5461, also known as Pidnarulex, is a First-in-Class anticancer drug that has received 'Fast Track Designation' approval from the FDA. Initially reported to inhibit Pol I-driven rDNA transcription, CX-5461 was recently identified as a G-quadruplex structure (G4) stabilizer and is currently completed or undergoing multiple Phase I clinical trials in patients with breast and ovarian cancers harboring BRCA1/2, PALB2, or other DNA repair deficiencies. Additionally, preclinical studies have confirmed that CX-5461 demonstrates promising therapeutic effects against multifarious non-cancer diseases, including viral infections, and autoimmune diseases. This review summarizes the mechanisms of CX-5461, including its transcriptional inhibition of rDNA, binding to G4, and toxicity towards topoisomerase, along with its research status and therapeutic effects across various diseases. Lastly, this review highlights the targeted therapy strategy of CX-5461 based on nanomedicine delivery, particularly the drug delivery utilizing the nucleic acid aptamer AS1411, which contains a G4 motif to specifically target the highly expressed nucleolin on the surface of tumor cell membranes; It also anticipates the strategy of coupling CX-5461 with peptide nucleic acids and locked nucleic acids to achieve dual targeting, thereby realizing individualized G4-targeting by CX-5461. This review aims to provide a general overview of the progress of CX-5461 in recent years and suggest potential strategies for disease treatment involving ribosomal RNA synthesis, G4, and topoisomerase.
Collapse
Affiliation(s)
- Hong-Xia Li
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, 646000, Sichuan, China
| | - Yi-Meng He
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jing Fei
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Man Guo
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, 646000, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, and Sichuan Clinical Research Center for Diabetes and Metabolic Disease, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chen Zeng
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Pi-Jun Yan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, 646000, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, and Sichuan Clinical Research Center for Diabetes and Metabolic Disease, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, 646000, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, and Sichuan Clinical Research Center for Diabetes and Metabolic Disease, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Gang Qin
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Fang-Yuan Teng
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, 646000, Sichuan, China.
- Sichuan Clinical Research Center for Nephropathy, and Sichuan Clinical Research Center for Diabetes and Metabolic Disease, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
27
|
Xie Q, Li K, Chen Y, Li Y, Jiang W, Cao W, Yu H, Fan D, Deng B. Gene therapy breakthroughs in ALS: a beacon of hope for 20% of ALS patients. Transl Neurodegener 2025; 14:19. [PMID: 40234983 PMCID: PMC12001736 DOI: 10.1186/s40035-025-00477-6] [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: 08/01/2024] [Accepted: 03/05/2025] [Indexed: 04/17/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease that remains incurable. Although the etiologies of ALS are diverse and the precise pathogenic mechanisms are not fully understood, approximately 20% of ALS cases are caused by genetic factors. Therefore, advancing targeted gene therapies holds significant promise, at least for the 20% of ALS patients with genetic etiologies. In this review, we summarize the main strategies and techniques of current ALS gene therapies based on ALS risk genes, and review recent findings from animal studies and clinical trials. Additionally, we highlight ALS-related genes with well-understood pathogenic mechanisms and the potential of numerous emerging gene-targeted therapeutic approaches for ALS.
Collapse
Affiliation(s)
- Qingjian Xie
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kezheng Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yinuo Chen
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yaojia Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
| | - Wenhua Jiang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China
| | - Wen Cao
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Huan Yu
- Department of Pediatrics, Second Affiliated Hospital and Yuying Children'S Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Binbin Deng
- Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 32500, China.
| |
Collapse
|
28
|
Zhang Z, Fu X, Wright N, Wang W, Ye Y, Asbury J, Li Y, Zhu C, Wu R, Wang S, Sun S. PTPσ-mediated PI3P regulation modulates neurodegeneration in C9ORF72-ALS/FTD. Neuron 2025; 113:1190-1205.e9. [PMID: 40073860 PMCID: PMC12005967 DOI: 10.1016/j.neuron.2025.02.005] [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: 07/16/2024] [Revised: 12/09/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025]
Abstract
The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the repeat expansion in C9ORF72. Dipeptide repeat (DPR) proteins translated from both sense and antisense repeats, especially arginine-rich DPRs (R-DPRs), contribute to neurodegeneration. Through CRISPR interference (CRISPRi) screening in human-derived neurons, we identified receptor-type tyrosine-protein phosphatase S (PTPσ) as a strong modifier of poly-GR-mediated toxicity. We showed that reducing PTPσ promotes the survival of both poly-GR- and poly-PR-expressing neurons by elevating phosphatidylinositol 3-phosphate (PI3P), accompanied by restored early endosomes and lysosomes. Remarkably, PTPσ knockdown or inhibition substantially rescues the PI3P-endolysosomal defects and improves the survival of C9ORF72-ALS/FTD patient-derived neurons. Furthermore, the PTPσ inhibitor diminishes GR toxicity and rescues pathological and behavioral phenotypes in mice. Overall, these findings emphasize the critical role of PI3P-mediated endolysosomal deficits induced by R-DPRs in disease pathogenesis and reveal the therapeutic potential of targeting PTPσ in C9ORF72-ALS/FTD.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xiujuan Fu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Noelle Wright
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Physiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Weiren Wang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biotechology Master Program, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yingzhi Ye
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Physiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Julie Asbury
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Notre Dame of Maryland University, Baltimore, MD 21210, USA
| | - Yini Li
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chengzhang Zhu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rong Wu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shaopeng Wang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shuying Sun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience and Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
29
|
Iyer K, Tenchov R, Sasso JM, Ralhan K, Jotshi J, Polshakov D, Maind A, Zhou QA. Rare Diseases, Spotlighting Amyotrophic Lateral Sclerosis, Huntington's Disease, and Myasthenia Gravis: Insights from Landscape Analysis of Current Research. Biochemistry 2025; 64:1698-1719. [PMID: 40169538 PMCID: PMC12004453 DOI: 10.1021/acs.biochem.4c00722] [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: 10/27/2024] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Rare diseases are a diverse group of disorders that, despite each individual condition's rarity, collectively affect a significant portion of the global population. Currently approximately 10,000 rare diseases exist globally, with 80% of these diseases being identified as having genetic origins. In this Review, we examine data from the CAS Content Collection to summarize scientific progress in the area of rare diseases. We examine the publication landscape in the area in an effort to provide insights into current advances and developments. We then discuss the evolution of key concepts in the field, genetic associations, as well as the major technologies and development pipelines of rare disease treatments. We focus our attention on three specific rare diseases: (i) amyotrophic lateral sclerosis, a terminal neurodegenerative disease affecting the central nervous system resulting in progressive loss of motor neurons that control voluntary muscles; (ii) Huntington's disease, another terminal neurodegenerative disease that causes progressive degeneration of nerve cells in the brain, with a wide impact on a person's functional abilities; and (iii) myasthenia gravis, a chronic autoimmune synaptopathy leading to skeletal muscle weakness. While the pathogenesis of these rare diseases is being elucidated, there is neither a cure nor preventative treatment available, only symptomatic treatment. The objective of the paper is to provide a broad overview of the evolving landscape of current knowledge on rare diseases and specifically on the biology and genetics of the three spotlighted diseases, to outline challenges and evaluate growth opportunities, an aim to further efforts in solving the remaining challenges.
Collapse
Affiliation(s)
- Kavita
A. Iyer
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Dmitrii Polshakov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Ankush Maind
- ACS
International India Pvt. Ltd., Pune 411044, India
| | | |
Collapse
|
30
|
Disney MD. The Druggable Transcriptome Project: From Chemical Probes to Precision Medicines. Biochemistry 2025; 64:1647-1661. [PMID: 40131857 PMCID: PMC12005196 DOI: 10.1021/acs.biochem.5c00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025]
Abstract
RNA presents abundant opportunities as a drug target, offering significant potential for small molecule medicine development. The transcriptome, comprising both coding and noncoding RNAs, is a rich area for therapeutic innovation, yet challenges persist in targeting RNA with small molecules. RNA structure can be predicted with or without experimental data, but discrepancies with the actual biological structure can impede progress. Prioritizing RNA targets supported by genetic or evolutionary evidence enhances success. Further, small molecules must demonstrate binding to RNA in cells, not solely in vitro, to validate both the target and compound. Effective small molecule binders modulate functional sites that influence RNA biology, as binding to nonfunctional sites requires recruiting effector mechanisms, for example degradation, to achieve therapeutic outcomes. Addressing these challenges is critical to unlocking RNA's vast potential for small molecule medicines, and a strategic framework is proposed to navigate this promising field, with a focus on targeting human RNAs.
Collapse
Affiliation(s)
- Matthew D. Disney
- Department
of Chemistry, The Herbert Wertheim UF Scripps
Institute for Biomedical Innovation and Technology, 130 Scripps Way, Jupiter, Florida 33458, United States
- Department
of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
31
|
González-Sánchez M, Ramírez-Expósito MJ, Martínez-Martos JM. Pathophysiology, Clinical Heterogeneity, and Therapeutic Advances in Amyotrophic Lateral Sclerosis: A Comprehensive Review of Molecular Mechanisms, Diagnostic Challenges, and Multidisciplinary Management Strategies. Life (Basel) 2025; 15:647. [PMID: 40283201 PMCID: PMC12029092 DOI: 10.3390/life15040647] [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: 03/09/2025] [Revised: 03/28/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the progressive degeneration of upper and lower motor neurons, leading to muscle atrophy, paralysis, and respiratory failure. This comprehensive review synthesizes the current knowledge on ALS pathophysiology, clinical heterogeneity, diagnostic frameworks, and evolving therapeutic strategies. Mechanistically, ALS arises from complex interactions between genetic mutations (e.g., in C9orf72, SOD1, TARDBP (TDP-43), and FUS) and dysregulated cellular pathways, including impaired RNA metabolism, protein misfolding, nucleocytoplasmic transport defects, and prion-like propagation of toxic aggregates. Phenotypic heterogeneity, manifesting as bulbar-, spinal-, or respiratory-onset variants, complicates its early diagnosis, which thus necessitates the rigorous application of the revised El Escorial criteria and emerging biomarkers such as neurofilament light chain. Clinically, ALS intersects with frontotemporal dementia (FTD) in up to 50% of the cases, driven by shared TDP-43 pathology and C9orf72 hexanucleotide expansions. Epidemiological studies have revealed a lifetime risk of 1:350, with male predominance (1.5:1) and peak onset between 50 and 70 years. Disease progression varies widely, with a median survival of 2-4 years post-diagnosis, underscoring the urgency for early intervention. Approved therapies, including riluzole (glutamate modulation), edaravone (antioxidant), and tofersen (antisense oligonucleotide), offer modest survival benefits, while dextromethorphan/quinidine alleviates the pseudobulbar affect. Non-pharmacological treatment advances, such as non-invasive ventilation (NIV), prolong survival by 13 months and improve quality of life, particularly in bulb-involved patients. Multidisciplinary care-integrating physical therapy, respiratory support, nutritional management, and cognitive assessments-is critical to addressing motor and non-motor symptoms (e.g., dysphagia, spasticity, sleep disturbances). Emerging therapies show promise in preclinical models. However, challenges persist in translating genetic insights into universally effective treatments. Ethical considerations, including euthanasia and end-of-life decision-making, further highlight the need for patient-centered communication and palliative strategies.
Collapse
Affiliation(s)
| | | | - José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, School of Health Sciences, University of Jaén, E23071 Jaén, Spain; (M.G.-S.); (M.J.R.-E.)
| |
Collapse
|
32
|
Zhang Y, Hulsman M, Salazar A, Tesi N, Knoop L, van der Lee S, Wijesekera S, Krizova J, Kamsteeg EJ, Holstege H. Multisample motif discovery and visualization for tandem repeats. Genome Res 2025; 35:850-862. [PMID: 39537359 PMCID: PMC12047238 DOI: 10.1101/gr.279278.124] [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: 03/08/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Tandem repeats (TRs) occupy a significant portion of the human genome and are a source of polymorphisms due to variations in sizes and motif compositions. Some of these variations have been associated with various neuropathological disorders, highlighting the clinical importance of assessing the motif structure of TRs. Moreover, assessing the TR motif variation can offer valuable insights into evolutionary dynamics and population structure. Previously, characterizations of TRs were limited by short-read sequencing technology, which lacks the ability to accurately capture the full TR sequences. As long-read sequencing becomes more accessible and can capture the full complexity of TRs, there is now also a need for tools to characterize and analyze TRs using long-read data across multiple samples. In this study, we present MotifScope, a novel algorithm for the characterization and visualization of TRs based on a de novo k-mer approach for motif discovery. Comparative analysis against established tools reveals that MotifScope can identify a greater number of motifs and more accurately represent the underlying repeat sequences. Moreover, MotifScope has been specifically designed to enable motif composition comparisons across assemblies of different individuals, as well as across long-read sequencing reads within an individual, through combined motif discovery and sequence alignment. We showcase potential applications of MotifScope in diverse fields, including population genetics, clinical settings, and forensic analyses.
Collapse
Affiliation(s)
- Yaran Zhang
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Marc Hulsman
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
| | - Alex Salazar
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Niccolò Tesi
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
| | - Lydian Knoop
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Sven van der Lee
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081HV Amsterdam, The Netherlands
| | - Sanduni Wijesekera
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Jana Krizova
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands
| | - Henne Holstege
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands;
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081HV Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
33
|
D'Anna L, Wragg D, Mauro D, Rubino S, Terenzi A, Barone G, Thomas SR, Casini A, Bonsignore R, Spinello A. Unraveling the Molecular Basis for G-Quadruplex-Binders to ALS/FTD-Associated G4C2 Repeats of the C9orf72 Gene. Chembiochem 2025; 26:e202400974. [PMID: 39670345 PMCID: PMC12007073 DOI: 10.1002/cbic.202400974] [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: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
The most recurrent familial cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the presence of an abnormal number of intronic GGGGCC (G4C2) repetitions in the C9orf72 gene, which has been proposed to drive ALS/FTD pathogenesis. Recently, it has been shown that such G4C2 repetitions can fold into G-quadruplex (G4) secondary structures. These G4s have been selectively stabilized by small-molecule binders, furnishing proof-of-principle that targeting these non-canonical nucleic acid sequences represents a novel and effective therapeutic strategy to tackle neurodegenerative disorders. However, precise information on the mechanism of action of these compounds is still lacking. Here, by performing in silico investigations, we unraveled the molecular basis for the selectivity of a series of known structurally related C9orf72 G4-binders. Moreover, we investigated the binding properties of a strong and selective metal-based G4 stabilizer, the AuI bis-N-heterocyclic carbene (NHC) complex - Au(TMX)2 - showing that it moderately stabilizes G4C2 G4 RNA by Förster resonance energy transfer (FRET) DNA melting assays. Using metadynamics (metaD) simulations, the Au(TMX)2 binding mode and the associated free-energy landscape were also evaluated. This information paves the way for developing improved compounds to tackle ALS/FTD neurodegenerative disorders.
Collapse
Affiliation(s)
- Luisa D'Anna
- Department of Biological, Chemical, and Pharmaceutical SciencesTechnologies, Università di PalermoViale delle Scienze Edificio 1790128PalermoItaly
| | - Darren Wragg
- Chair of Medicinal and Bioinorganic ChemistrySchool of Natural SciencesDepartment of ChemistryTechnical University of Munich (TUM)Lichtenbergstr. 485748Garching b. MünchenGermany
| | - Daniela Mauro
- Department of Biological, Chemical, and Pharmaceutical SciencesTechnologies, Università di PalermoViale delle Scienze Edificio 1790128PalermoItaly
| | - Simona Rubino
- Department of Biological, Chemical, and Pharmaceutical SciencesTechnologies, Università di PalermoViale delle Scienze Edificio 1790128PalermoItaly
| | - Alessio Terenzi
- Department of Biological, Chemical, and Pharmaceutical SciencesTechnologies, Università di PalermoViale delle Scienze Edificio 1790128PalermoItaly
| | - Giampaolo Barone
- Department of Biological, Chemical, and Pharmaceutical SciencesTechnologies, Università di PalermoViale delle Scienze Edificio 1790128PalermoItaly
| | - Sophie R. Thomas
- Department of Inorganic ChemistryUniversity of ViennaWähringer Straße. 42ViennaAustria
| | - Angela Casini
- Chair of Medicinal and Bioinorganic ChemistrySchool of Natural SciencesDepartment of ChemistryTechnical University of Munich (TUM)Lichtenbergstr. 485748Garching b. MünchenGermany
| | - Riccardo Bonsignore
- Department of Biological, Chemical, and Pharmaceutical SciencesTechnologies, Università di PalermoViale delle Scienze Edificio 1790128PalermoItaly
| | - Angelo Spinello
- Department of Biological, Chemical, and Pharmaceutical SciencesTechnologies, Università di PalermoViale delle Scienze Edificio 1790128PalermoItaly
| |
Collapse
|
34
|
Luteijn MJ, Bhaskar V, Trojer D, Schürz M, Mahboubi H, Handl C, Pizzato N, Pfeifer M, Dafinca R, Voshol H, Giorgetti E, Manneville C, Garnier IPM, Müller M, Zeng F, Buntin K, Markwalder R, Schröder H, Weiler J, Khar D, Schuhmann T, Groot-Kormelink PJ, Keller CG, Farmer P, MacKay A, Beibel M, Roma G, D’Ario G, Merkl C, Schebesta M, Hild M, Elwood F, Vahsen BF, Ripin N, Clery A, Allain F, Labow M, Gabriel D, Chao JA, Talbot K, Nash M, Hunziker J, Meisner-Kober NC. High-throughput screen of 100 000 small molecules in C9ORF72 ALS neurons identifies spliceosome modulators that mobilize G4C2 repeat RNA into nuclear export and repeat associated non-canonical translation. Nucleic Acids Res 2025; 53:gkaf253. [PMID: 40207633 PMCID: PMC11983130 DOI: 10.1093/nar/gkaf253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025] Open
Abstract
An intronic G4C2 repeat expansion in the C9ORF72 gene is the major known cause for Amyotrophic Lateral Sclerosis (ALS), with current evidence for both, loss of function and pathological gain of function disease mechanisms. We screened 96 200 small molecules in C9ORF72 patient iPS neurons for modulation of nuclear G4C2 RNA foci and identified 82 validated hits, including the Brd4 inhibitor JQ1 as well as novel analogs of Spliceostatin-A, a known modulator of SF3B1, the branch point binding protein of the U2-snRNP. Spliceosome modulation by these SF3B1 targeted compounds recruits SRSF1 to nuclear G4C2 RNA, mobilizing it from RNA foci into nucleocytoplasmic export. This leads to increased repeat-associated non-canonical (RAN) translation and ultimately, enhanced cell toxicity. Our data (i) provide a new pharmacological entry point with novel as well as known, publicly available tool compounds for dissection of C9ORF72 pathobiology in C9ORF72 ALS models, (ii) allowing to differentially modulate RNA foci versus RAN translation, and (iii) suggest that therapeutic RNA foci elimination strategies warrant caution due to a potential storage function, counteracting translation into toxic dipeptide repeat polyproteins. Instead, our data support modulation of nuclear export via SRSF1 or SR protein kinases as possible targets for future pharmacological drug discovery.
Collapse
Affiliation(s)
- Maartje J Luteijn
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Varun Bhaskar
- Friedrich Miescher Institute for Biomedical Research, Department Genomic Regulation, Basel, 4056, Switzerland
| | - Dominic Trojer
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Melanie Schürz
- Paris-Lodron University of Salzburg, Department of Biosciences and Medical Biology,, Salzburg, 5020,Austria
- Ludwig Boltzmann Institute for Nanovesicular Precision Medicine at the Paris Lodron University Salzburg, 5020, Austria
| | - Hicham Mahboubi
- Friedrich Miescher Institute for Biomedical Research, Department Genomic Regulation, Basel, 4056, Switzerland
| | - Cornelia Handl
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Nicolas Pizzato
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Martin Pfeifer
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Ruxandra Dafinca
- University of Oxford, John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, Oxford, OX3 9DU, United Kingdom
| | - Hans Voshol
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Elisa Giorgetti
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Carole Manneville
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Isabelle P M Garnier
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Matthias Müller
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Fanning Zeng
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Kathrin Buntin
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Roger Markwalder
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Harald Schröder
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Jan Weiler
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Dora Khar
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Tim Schuhmann
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Paul J Groot-Kormelink
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Caroline Gubser Keller
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Pierre Farmer
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Angela MacKay
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Martin Beibel
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Guglielmo Roma
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Giovanni D’Ario
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Claudia Merkl
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Michael Schebesta
- Novartis Institutes for Biomedical Research, Department Discovery Sciences, Cambridge, MA02139, United States
| | - Marc Hild
- Novartis Institutes for Biomedical Research, Department Discovery Sciences, Cambridge, MA02139, United States
| | - Fiona Elwood
- Novartis Institutes for Biomedical Research, Department Discovery Sciences, Cambridge, MA02139, United States
| | - Björn F Vahsen
- ETH Zürich, Department of Biology, Institute f. Molekularbiol.u.Biophysik, Zürich, 8093, Switzerland
| | - Nina Ripin
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
- ETH Zürich, Department of Biology, Institute f. Molekularbiol.u.Biophysik, Zürich, 8093, Switzerland
| | - Antoine Clery
- ETH Zürich, Department of Biology, Institute f. Molekularbiol.u.Biophysik, Zürich, 8093, Switzerland
| | - Frederic Allain
- ETH Zürich, Department of Biology, Institute f. Molekularbiol.u.Biophysik, Zürich, 8093, Switzerland
| | - Mark Labow
- Novartis Institutes for Biomedical Research, Department Discovery Sciences, Cambridge, MA02139, United States
| | - Daniela Gabriel
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Jeffrey A Chao
- Friedrich Miescher Institute for Biomedical Research, Department Genomic Regulation, Basel, 4056, Switzerland
| | - Kevin Talbot
- University of Oxford, John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, Oxford, OX3 9DU, United Kingdom
| | - Mark Nash
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Jürg Hunziker
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
| | - Nicole C Meisner-Kober
- Novartis Institutes for Biomedical Research, Department Global Discovery Chemistry, Basel, 4056, Switzerland
- Paris-Lodron University of Salzburg, Department of Biosciences and Medical Biology,, Salzburg, 5020,Austria
- Ludwig Boltzmann Institute for Nanovesicular Precision Medicine at the Paris Lodron University Salzburg, 5020, Austria
| |
Collapse
|
35
|
Ms S, Banerjee S, D'Mello SR, Dastidar SG. Amyotrophic Lateral Sclerosis: Focus on Cytoplasmic Trafficking and Proteostasis. Mol Neurobiol 2025:10.1007/s12035-025-04831-7. [PMID: 40180687 DOI: 10.1007/s12035-025-04831-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/09/2025] [Indexed: 04/05/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal motor neuron disease characterized by the pathological loss of upper and lower motor neurons. Whereas most ALS cases are caused by a combination of environmental factors and genetic susceptibility, in a relatively small proportion of cases, the disorder results from mutations in genes that are inherited. Defects in several different cellular mechanisms and processes contribute to the selective loss of motor neurons (MNs) in ALS. Prominent among these is the accumulation of aggregates of misfolded proteins or peptides which are toxic to motor neurons. These accumulating aggregates stress the ability of the endoplasmic reticulum (ER) to function normally, cause defects in the transport of proteins between the ER and Golgi, and impair the transport of RNA, proteins, and organelles, such as mitochondria, within axons and dendrites, all of which contribute to the degeneration of MNs. Although dysfunction of a variety of cellular processes combines towards the pathogenesis of ALS, in this review, we focus on recent advances concerning the involvement of defective ER stress, vesicular transport between the ER and Golgi, and axonal transport.
Collapse
Affiliation(s)
- Shrilaxmi Ms
- Center for Molecular Neuroscience, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Saradindu Banerjee
- Center for Molecular Neuroscience, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Santosh R D'Mello
- Center for Molecular Neuroscience, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
- College of Arts and Sciences, Louisiana State University, Shreveport, LA, 71115, USA.
| | - Somasish Ghosh Dastidar
- Center for Molecular Neuroscience, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
36
|
Schierbaum L, Quiroz V, Yang K, Rong J, Battaglia N, Zubair U, Christie M, Davis M, Calame D, Danzi MC, Finkel RS, Burns J, Gilbert DL, Mingbunjerdsuk D, Pruitt G, Pruitt N, Cobb J, Sadjadi R, Cashman CR, Blackstone C, Fink JK, Shy ME, Zuchner S, Ebrahimi-Fakhari D. The Spastic Paraplegia-Centers of Excellence Research Network (SP-CERN): Clinical Trial Readiness for Hereditary Spastic Paraplegia. Neurol Genet 2025; 11:e200249. [PMID: 39996129 PMCID: PMC11849523 DOI: 10.1212/nxg.0000000000200249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025]
Abstract
Objectives The primary objective of this paper was to present the establishment of the Spastic Paraplegia-Centers of Excellence Research Network (SP-CERN) aimed at promoting clinical trial readiness for hereditary spastic paraplegia (HSP). SP-CERN is unique in its approach to addressing the diagnostic and therapeutic challenges associated with HSP through a large-scale, collaborative effort. Methods Participants with HSP are identified through multicenter collaborations across 11 institutions in the United States. SP-CERN systematically collects longitudinal clinical data, biospecimens, and wearable device data from patients. Data are stored in a centralized REDCap database, facilitating shared access for analysis. Patients are evaluated using standardized assessment tools for motor function, biomarkers, and digital outcome measures. Results SP-CERN has established a biorepository, centralized data collection methods, and standardized clinical assessments. It is conducting natural history studies for all HSP subtypes, enabling the validation of biomarkers and development of gene-based therapies. Discussion SP-CERN's collaborative approach bridges gaps in clinical care and research for HSP by improving diagnostic capabilities and promoting clinical trial readiness. This initiative represents a framework for rare disease research, accelerating the development of novel therapies and improving patient outcomes through standardized, multi-institutional collaboration.
Collapse
Affiliation(s)
- Luca Schierbaum
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital
| | - Vicente Quiroz
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital
| | - Kathryn Yang
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital
| | - Joshua Rong
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital
| | - Nicole Battaglia
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital
| | - Umar Zubair
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital
| | - Michelle Christie
- Division of Neurology and Rehabilitation Medicine, Scottish Rite Hospital for Children
| | - Marie Davis
- Department of Neurology, University of Washington
| | - Daniel Calame
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine
| | - Matt C Danzi
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine
| | - Richard S Finkel
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children's Research Hospital
| | - Joshua Burns
- Disability Prevention Program, Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital
| | - Donald L Gilbert
- Division of Neurology, Cincinnati Children's Hospital Medical Center
| | - Dararat Mingbunjerdsuk
- Division of Pediatric Neurology, Department of Neurology, Seattle Children's Hospital, University of Washington
| | | | | | | | - Reza Sadjadi
- Department of Neurology, Massachusetts General Hospital
| | | | | | - John K Fink
- Department of Neurology, University of Michigan; and
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa
| | - Stephan Zuchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine
| | | |
Collapse
|
37
|
Javadzadeh S, Adamson A, Park J, Jo SY, Ding YC, Bakhtiari M, Bansal V, Neuhausen SL, Bafna V. Analysis of targeted and whole genome sequencing of PacBio HiFi reads for a comprehensive genotyping of gene-proximal and phenotype-associated Variable Number Tandem Repeats. PLoS Comput Biol 2025; 21:e1012885. [PMID: 40193344 PMCID: PMC11975116 DOI: 10.1371/journal.pcbi.1012885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/17/2025] [Indexed: 04/09/2025] Open
Abstract
Variable Number Tandem repeats (VNTRs) refer to repeating motifs of size greater than five bp. VNTRs are an important source of genetic variation, and have been associated with multiple Mendelian and complex phenotypes. However, the highly repetitive structures require reads to span the region for accurate genotyping. Pacific Biosciences HiFi sequencing spans large regions and is highly accurate but relatively expensive. Therefore, targeted sequencing approaches coupled with long-read sequencing have been proposed to improve efficiency and throughput. In this paper, we systematically explored the trade-off between targeted and whole genome HiFi sequencing for genotyping VNTRs. We curated a set of 10 , 787 gene-proximal (G-)VNTRs, and 48 phenotype-associated (P-)VNTRs of interest. Illumina reads only spanned 46% of the G-VNTRs and 71% of P-VNTRs, motivating the use of HiFi sequencing. We performed targeted sequencing with hybridization by designing custom probes for 9,999 VNTRs and sequenced 8 samples using HiFi and Illumina sequencing, followed by adVNTR genotyping. We compared these results against HiFi whole genome sequencing (WGS) data from 28 samples in the Human Pangenome Reference Consortium (HPRC). With the targeted approach only 4,091 (41%) G-VNTRs and only 4 (8%) of P-VNTRs were spanned with at least 15 reads. A smaller subset of 3,579 (36%) G-VNTRs had higher median coverage of at least 63 spanning reads. The spanning behavior was consistent across all 8 samples. Among 5,638 VNTRs with low-coverage ( < 15), 67% were located within GC-rich regions ( > 60%). In contrast, the 40X WGS HiFi dataset spanned 98% of all VNTRs and 49 (98%) of P-VNTRs with at least 15 spanning reads, albeit with lower coverage. Spanning reads were sufficient for accurate genotyping in both cases. Our findings demonstrate that targeted sequencing provides consistently high coverage for a small subset of low-GC VNTRs, but WGS is more effective for broad and sufficient sampling of a large number of VNTRs.
Collapse
Affiliation(s)
- Sara Javadzadeh
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Aaron Adamson
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Jonghun Park
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Se-Young Jo
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Yuan-Chun Ding
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Mehrdad Bakhtiari
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Vikas Bansal
- School of Medicine, University of California, San Diego La Jolla, California, United States of America
| | - Susan L. Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
38
|
van der Schaar J, van der Lee SJ, Asscher ECA, Pijnenburg YAL, de Geus CM, Bredenoord AL, van der Flier WM, van den Hoven MA, Smets EMA, Visser LNC. Deciding on genetic testing for familial dementia: Perspectives of patients and families. Alzheimers Dement 2025; 21:e70140. [PMID: 40189825 PMCID: PMC11972981 DOI: 10.1002/alz.70140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION We explored patients' and families' interest in, predictors of, and considerations regarding genetic testing for monogenic causes of dementia in a diagnostic setting. METHODS This mixed-methods study evaluated 519 consecutive Alzheimer Center Amsterdam patients for monogenic testing eligibility. Among those qualifying, differences between testers and non-testers were analyzed. Thirty-three patients completed questionnaires. Additionally, we conducted 21 semi-structured interviews with 15 patients and 18 relatives. Verbatim transcripts were analyzed inductively. RESULTS Of 138 (27%) eligible patients (46% female, age 61 ± 8 years, Mini-Mental State Examination [MMSE] 22 ± 6), 75 (54%) underwent genetic testing. Testers had better cognition, higher quality of life, and more often undetermined diagnoses than non-testers (all p < 0.05). Decisions were guided by intuitive, value-driven judgments: testers sought to provide heredity information to relatives, enhance actionability, and reduce uncertainty, while non-testers worried about psychosocial impact on family, or unfavorable timing. DISCUSSION The substantial interest in genetic testing for monogenic causes of dementia underscores the need for further research into the implications of disclosing test results to memory clinic patients. HIGHLIGHTS Half of memory clinic patients' who met eligibility criteria proceeded with genetic testing. Those tested were more likely to have an undetermined diagnosis, better cognition, and higher quality of life. Decisions were motivated less by deliberation of factual information, and more by quick, intuitive judgments. Motivations pro included providing information, enhancing actionability, and resolving uncertainty. Motivations con comprised concerns about the emotional burden and disruptive impact on their family.
Collapse
Affiliation(s)
- Jetske van der Schaar
- Section Genomics of Neurodegenerative Diseases and AgingDepartment of Human GeneticsVrije Universiteit AmsterdamAmsterdamNetherlands
- Alzheimer Center AmsterdamNeurology, Vrije Universiteit AmsterdamAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationResearch & Diagnostics Center (RDC) ‐ ADOREAmsterdamThe Netherlands
| | - Sven J. van der Lee
- Section Genomics of Neurodegenerative Diseases and AgingDepartment of Human GeneticsVrije Universiteit AmsterdamAmsterdamNetherlands
- Alzheimer Center AmsterdamNeurology, Vrije Universiteit AmsterdamAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationResearch & Diagnostics Center (RDC) ‐ ADOREAmsterdamThe Netherlands
| | | | - Yolande A. L. Pijnenburg
- Alzheimer Center AmsterdamNeurology, Vrije Universiteit AmsterdamAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationResearch & Diagnostics Center (RDC) ‐ ADOREAmsterdamThe Netherlands
| | - Christa M. de Geus
- Clinical GeneticsDepartment of Human GeneticsVrije Universiteit AmsterdamAmsterdamNetherlands
| | | | - Wiesje M. van der Flier
- Alzheimer Center AmsterdamNeurology, Vrije Universiteit AmsterdamAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationResearch & Diagnostics Center (RDC) ‐ ADOREAmsterdamThe Netherlands
- Department of Epidemiology & Data sciencesVrije Universiteit AmsterdamAmsterdam UMC location VUmcAmsterdamThe Netherlands
| | | | - Ellen M. A. Smets
- Department of Medical PsychologyAmsterdam UMClocation AMCAmsterdamThe Netherlands
- Amsterdam Public HealthQuality of CareAmsterdamThe Netherlands
| | - Leonie N. C. Visser
- Alzheimer Center AmsterdamNeurology, Vrije Universiteit AmsterdamAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationResearch & Diagnostics Center (RDC) ‐ ADOREAmsterdamThe Netherlands
- Department of Medical PsychologyAmsterdam UMClocation AMCAmsterdamThe Netherlands
- Amsterdam Public HealthQuality of CareAmsterdamThe Netherlands
- Department of Bioethics and Health HumanitiesJulius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
39
|
Askarova A, Yaa RM, Marzi SJ, Nott A. Genetic risk for neurodegenerative conditions is linked to disease-specific microglial pathways. PLoS Genet 2025; 21:e1011407. [PMID: 40202986 PMCID: PMC12017514 DOI: 10.1371/journal.pgen.1011407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 04/23/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Genome-wide association studies have identified thousands of common variants associated with an increased risk of neurodegenerative disorders. However, the noncoding localization of these variants has made the assignment of target genes for brain cell types challenging. Genomic approaches that infer chromosomal 3D architecture can link noncoding risk variants and distal gene regulatory elements such as enhancers to gene promoters. By using enhancer-to-promoter interactome maps for human microglia, neurons, and oligodendrocytes, we identified cell-type-specific enrichment of genetic heritability for brain disorders through stratified linkage disequilibrium score regression. Our analysis suggests that genetic heritability for multiple neurodegenerative disorders is enriched at microglial chromatin contact sites, while schizophrenia heritability is predominantly enriched at chromatin contact sites in neurons followed by oligodendrocytes. Through Hi-C coupled multimarker analysis of genomic annotation (H-MAGMA), we identified disease risk genes for Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis and schizophrenia. We found that disease-risk genes were overrepresented in microglia compared to other brain cell types across neurodegenerative conditions and within neurons for schizophrenia. Notably, the microglial risk genes and pathways identified were largely specific to each disease. Our findings reinforce microglia as an important, genetically informed cell type for therapeutic interventions in neurodegenerative conditions and highlight potentially targetable disease-relevant pathways.
Collapse
Affiliation(s)
- Aydan Askarova
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- United Kingdom Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Reuben M. Yaa
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- United Kingdom Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Sarah J. Marzi
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- United Kingdom Dementia Research Institute, King’s College London, London, United Kingdom
| | - Alexi Nott
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- United Kingdom Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
40
|
Giblin A, Cammack AJ, Blomberg N, Anoar S, Mikheenko A, Carcolé M, Atilano ML, Hull A, Shen D, Wei X, Coneys R, Zhou L, Mohammed Y, Olivier-Jimenez D, Wang LY, Kinghorn KJ, Niccoli T, Coyne AN, van der Kant R, Lashley T, Giera M, Partridge L, Isaacs AM. Neuronal polyunsaturated fatty acids are protective in ALS/FTD. Nat Neurosci 2025; 28:737-747. [PMID: 40000803 PMCID: PMC11976277 DOI: 10.1038/s41593-025-01889-3] [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: 01/22/2024] [Accepted: 01/07/2025] [Indexed: 02/27/2025]
Abstract
Here we report a conserved transcriptomic signature of reduced fatty acid and lipid metabolism gene expression in a Drosophila model of C9orf72 repeat expansion, the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), and in human postmortem ALS spinal cord. We performed lipidomics on C9 ALS/FTD Drosophila, induced pluripotent stem (iPS) cell neurons and postmortem FTD brain tissue. This revealed a common and specific reduction in phospholipid species containing polyunsaturated fatty acids (PUFAs). Feeding C9 ALS/FTD flies PUFAs yielded a modest increase in survival. However, increasing PUFA levels specifically in neurons of C9 ALS/FTD flies, by overexpressing fatty acid desaturase enzymes, led to a substantial extension of lifespan. Neuronal overexpression of fatty acid desaturases also suppressed stressor-induced neuronal death in iPS cell neurons of patients with both C9 and TDP-43 ALS/FTD. These data implicate neuronal fatty acid saturation in the pathogenesis of ALS/FTD and suggest that interventions to increase neuronal PUFA levels may be beneficial.
Collapse
Affiliation(s)
- Ashling Giblin
- UK Dementia Research Institute, UCL, London, UK
- Institute of Healthy Ageing, UCL, London, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alexander J Cammack
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Niek Blomberg
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Alla Mikheenko
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Mireia Carcolé
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | - Alex Hull
- Institute of Healthy Ageing, UCL, London, UK
| | - Dunxin Shen
- Institute of Healthy Ageing, UCL, London, UK
| | - Xiaoya Wei
- Institute of Healthy Ageing, UCL, London, UK
| | - Rachel Coneys
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Lele Zhou
- UK Dementia Research Institute, UCL, London, UK
- Institute of Healthy Ageing, UCL, London, UK
| | - Yassene Mohammed
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Damien Olivier-Jimenez
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Lian Y Wang
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Alyssa N Coyne
- Department of Neurology, Johns Hopkins University, Baltimore, MA, USA
- Brain Science Institute, Johns Hopkins University, Baltimore, MA, USA
| | - Rik van der Kant
- Alzheimer Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Martin Giera
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Adrian M Isaacs
- UK Dementia Research Institute, UCL, London, UK.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
41
|
Liu Y, Xia K. Aberrant Short Tandem Repeats: Pathogenicity, Mechanisms, Detection, and Roles in Neuropsychiatric Disorders. Genes (Basel) 2025; 16:406. [PMID: 40282366 PMCID: PMC12026680 DOI: 10.3390/genes16040406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
Short tandem repeat (STR) sequences are highly variable DNA segments that significantly contribute to human neurodegenerative disorders, highlighting their crucial role in neuropsychiatric conditions. This article examines the pathogenicity of abnormal STRs and classifies tandem repeat expansion disorders(TREDs), emphasizing their genetic characteristics, mechanisms of action, detection methods, and associated animal models. STR expansions exhibit complex genetic patterns that affect the age of onset and symptom severity. These expansions disrupt gene function through mechanisms such as gene silencing, toxic gain-of-function mutations leading to RNA and protein toxicity, and the generation of toxic peptides via repeat-associated non-AUG (RAN) translation. Advances in sequencing technologies-from traditional PCR and Southern blotting to next-generation and long-read sequencing-have enhanced the accuracy of STR variation detection. Research utilizing these technologies has linked STR expansions to a range of neuropsychiatric disorders, including autism spectrum disorders and schizophrenia, highlighting their contribution to disease risk and phenotypic expression through effects on genes involved in neurodevelopment, synaptic function, and neuronal signaling. Therefore, further investigation is essential to elucidate the intricate interplay between STRs and neuropsychiatric diseases, paving the way for improved diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yuzhong Liu
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China;
- MOE Key Lab of Rare Pediatric Diseases, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Kun Xia
- Institute of Cytology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China;
- MOE Key Lab of Rare Pediatric Diseases, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| |
Collapse
|
42
|
Xu R, Kang Q, Yang X, Yi P, Zhang R. Unraveling Molecular Targets for Neurodegenerative Diseases Through Caenorhabditis elegans Models. Int J Mol Sci 2025; 26:3030. [PMID: 40243699 PMCID: PMC11988803 DOI: 10.3390/ijms26073030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and prion disease, represent a group of age-related disorders that pose a growing and formidable challenge to global health. Despite decades of extensive research that has uncovered key genetic factors and biochemical pathways, the precise molecular mechanisms underlying these diseases and effective therapeutic strategies remain elusive. Caenorhabditis elegans (C. elegans) has emerged as a powerful model organism for studying NDDs due to its unique biological features such as genetic tractability, conserved molecular pathways, and ease of high-throughput screening. This model provides an exceptional platform for identifying molecular targets associated with NDDs and developing novel therapeutic interventions. This review highlights the critical role of C. elegans in elucidating the complex molecular mechanisms of human NDDs, with a particular focus on recent advancements and its indispensable contributions to the discovery of molecular targets and therapeutic strategies for these NDDs.
Collapse
Affiliation(s)
- Rongmei Xu
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei 230002, China; (R.X.); (X.Y.)
| | - Qiaoju Kang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (Q.K.); (P.Y.)
| | - Xuefei Yang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei 230002, China; (R.X.); (X.Y.)
| | - Ping Yi
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (Q.K.); (P.Y.)
| | - Rongying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (Q.K.); (P.Y.)
| |
Collapse
|
43
|
Vaughan DP, Real R, Jensen MT, Fumi RG, Hodgson M, Jabbari E, Lux D, Wu L, Warner TT, Jaunmuktane Z, Revesz T, Rowe JB, Rohrer J, Morris HR. Analysis of C9orf72 repeat length in progressive supranuclear palsy, corticobasal syndrome, corticobasal degeneration, and atypical parkinsonism. J Neurol 2025; 272:293. [PMID: 40138021 PMCID: PMC11947049 DOI: 10.1007/s00415-025-12990-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/09/2025] [Accepted: 02/15/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Pathogenic hexanucleotide repeat expansions in C9orf72 are the commonest genetic cause of frontotemporal dementia and/or amyotrophic lateral sclerosis. There is growing interest in intermediate repeat expansions in C9orf72 and their relationship to a wide range of neurological presentations, including Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, corticobasal degeneration, and corticobasal syndromes. AIMS To assess the prevalence of intermediate C9orf72 repeat expansions in a large cohort of prospectively-recruited patients clinically diagnosed with progressive supranuclear palsy (PSP), corticobasal syndrome (CBS), and atypical parkinsonism (APS), compared with healthy controls. We also sought to replicate the association between C9orf72 repeat length and CBD in neuropathologically confirmed cases. METHODS 626 cases, including PSP (n = 366), CBS (n = 130), and APS (n = 53) from the PROSPECT study, and 77 cases with pathologically confirmed CBD were screened for intermediate repeat expansions in C9orf72 using repeat-primed PCR. These were compared to controls from the PROSPECT-M-UK study and from the 1958 Birth Cohort. RESULTS There was no difference in the mean or largest allele size in any affected patient group compared with controls. A higher proportion of our affected cohort had large C9orf72 repeat expansions compared to controls, but there was no difference when comparing the frequency of intermediate expansions between affected patients and controls. There was no relationship between repeat length and age at onset, level of disability, or survival. CONCLUSIONS Intermediate expansions in C9orf72 do not appear to be a genetic risk factor for PSP, CBS, CBD, or atypical parkinsonism. They are not associated with age at onset, disability, or survival in our study.
Collapse
Affiliation(s)
- David P Vaughan
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Marte Theilmann Jensen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Riona G Fumi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Megan Hodgson
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Edwin Jabbari
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Danielle Lux
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Lesley Wu
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Zane Jaunmuktane
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
- Queen Square Brain Bank, Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
| | - Tamas Revesz
- Queen Square Brain Bank, Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Jonathan Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK.
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
44
|
Wang KS, Smeyers J, Eggan K, Budnik B, Mordes DA. C9ORF72 poly-PR disrupts expression of ALS/FTD-implicated STMN2 through SRSF7. Acta Neuropathol Commun 2025; 13:67. [PMID: 40140908 PMCID: PMC11948778 DOI: 10.1186/s40478-025-01977-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
A hexanucleotide repeat expansion in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and combined ALS/FTD. The repeat is transcribed in the sense and the antisense directions to produce several dipeptide repeat proteins (DPRs) that have toxic gain-of-function effects; however, the mechanisms by which DPRs lead to neural dysfunction remain unresolved. Here, we observed that poly-proline-arginine (poly-PR) was sufficient to inhibit axonal regeneration of human induced pluripotent stem cell (iPSC)-derived neurons. Global phospho-proteomics revealed that poly-PR selectively perturbs nuclear RNA binding proteins (RBPs). In neurons, we found that depletion of one of these RBPs, SRSF7 (serine/arginine-rich splicing factor 7), resulted in decreased abundance of STMN2 (stathmin-2), though not TDP-43. STMN2 supports axon maintenance and repair and has been recently implicated in the pathogenesis of ALS/FTD. We observed that depletion of SRSF7 impaired axonal regeneration, a phenotype that could be rescued by exogenous STMN2. We propose that antisense repeat-encoded poly-PR perturbs RBPs, particularly SRSF7, resulting in reduced STMN2 and axonal repair defects in neurons. Hence, we provide a potential link between DPRs gain-of-function effects and STMN2 loss-of-function phenotypes in neurodegeneration.
Collapse
Affiliation(s)
- Karen S Wang
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Julie Smeyers
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bogdan Budnik
- Mass Spectrometry and Proteomic Laboratory, FAS Division of Science, Harvard University, Cambridge, MA, USA
- Wyss Institute, Harvard University, Boston, MA, USA
| | - Daniel A Mordes
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, CA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
45
|
Ramesh N, Evans A, Wojta K, Yang Z, Boks MM, Kahn RS, de Boer SCM, van der Lee SJ, Pijnenburg YAL, Reus LM, Ophoff RA. Accurate DNA Methylation Predictor for C9orf72 Repeat Expansion Alleles in the Pathogenic Range. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.643775. [PMID: 40196659 PMCID: PMC11974722 DOI: 10.1101/2025.03.20.643775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The hexanucleotide (G 4 C 2 ) repeat expansion in the promoter region of C9orf72 is the most frequent genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). In this study, we conducted a genome-wide DNA methylation (DNAm) analysis using EPIC version 2 (EPICv2) arrays on an FTD cohort comprising 27 carriers and 250 non-carriers of the pathogenic C9orf72 repeat expansion from the Amsterdam Dementia Cohort. We identified differentially methylated CpGs probes associated with the pathogenic C9orf72 expansion and used these findings to create a DNAm Least Absolute Shrinkage and Selection Operator (LASSO) predictor to identify repeat expansion carriers. Eight CpG sites at the C9orf72 locus were significantly differentially hypermethylated in repeat expansion carriers compared to non-carriers. The LASSO model predicted repeat expansion status with an average accuracy of 98.6%. The LASSO predictor was further validated in an independent cohort of 2,548 subjects with available EPICv2 data, identifying four C9orf72 repeat expansion carriers, subsequently confirmed by repeat-primed PCR. This result not only illustrates the accuracy of the DNAm predictor of C9orf72 repeat expansion carriers but also suggests that repeat expansion carriers may be more prevalent than expected. The identification of a highly accurate DNAm biomarker for a repeat expansion locus associated with neurodegenerative disorders may provide great value for studying this locus. The approach holds significant promise for investigating this and other repeat expansion loci, particularly given the growing interest in epigenetic epidemiological studies involving large cohorts with available DNAm data. Graphical abstract optional
Collapse
|
46
|
Scarcella S, Brambilla L, Quetti L, Rizzuti M, Melzi V, Galli N, Sali L, Costamagna G, Comi GP, Corti S, Gagliardi D. Unveiling amyotrophic lateral sclerosis complexity: insights from proteomics, metabolomics and microbiomics. Brain Commun 2025; 7:fcaf114. [PMID: 40161216 PMCID: PMC11952287 DOI: 10.1093/braincomms/fcaf114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Amyotrophic lateral sclerosis is the most common motor neuron disease and manifests as a clinically and genetically heterogeneous neurodegenerative disorder mainly affecting the motor systems. To date, despite promising results and accumulating knowledge on the pathomechanisms of amyotrophic lateral sclerosis, a specific disease-modifying treatment is still not available. In vitro and in vivo disease models coupled with multiomics techniques have helped elucidate the pathomechanisms underlying this disease. In particular, omics approaches are powerful tools for identifying new potential disease biomarkers that may be particularly useful for diagnosis, prognosis and assessment of treatment response. In turn, these findings could support physicians in stratifying patients into clinically relevant subgroups for the identification of the best therapeutic targets. Here, we provide a comprehensive review of the most relevant literature highlighting the importance of proteomics approaches in determining the role of pathogenic misfolded/aggregated proteins and the molecular mechanisms involved in the pathogenesis and progression of amyotrophic lateral sclerosis. In addition, we explored new findings arising from metabolomic and lipidomic studies, which can aid to elucidate the intricate metabolic alterations underlying amyotrophic lateral sclerosis pathology. Moreover, we integrated these insights with microbiomics data, providing a thorough understanding of the interplay between metabolic dysregulation and microbial dynamics in disease progression. Indeed, a greater integration of these multiomics data could lead to a deeper understanding of disease mechanisms, supporting the development of specific therapies for amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Simone Scarcella
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Università degli Studi di Milano, 20122 Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Noemi Galli
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Università degli Studi di Milano, 20122 Milan, Italy
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Gianluca Costamagna
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Università degli Studi di Milano, 20122 Milan, Italy
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giacomo Pietro Comi
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Università degli Studi di Milano, 20122 Milan, Italy
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefania Corti
- Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Università degli Studi di Milano, 20122 Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Delia Gagliardi
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
47
|
Zhou Z, Luquette LJ, Dong G, Kim J, Ku J, Kim K, Bae M, Shao DD, Sahile B, Miller MB, Huang AY, Nathan WJ, Nussenzweig A, Park PJ, Lagier-Tourenne C, Lee EA, Walsh CA. Recurrent patterns of widespread neuronal genomic damage shared by major neurodegenerative disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.641186. [PMID: 40093130 PMCID: PMC11908196 DOI: 10.1101/2025.03.03.641186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Alzheimer's disease (AD) are common neurodegenerative disorders for which the mechanisms driving neuronal death remain unclear. Single-cell whole-genome sequencing of 429 neurons from three C9ORF72 ALS, six C9ORF72 FTD, seven AD, and twenty-three neurotypical control brains revealed significantly increased burdens in somatic single nucleotide variant (sSNV) and insertion/deletion (sIndel) in all three disease conditions. Mutational signature analysis identified a disease-associated sSNV signature suggestive of oxidative damage and an sIndel process, affecting 28% of ALS, 79% of FTD, and 65% of AD neurons but only 5% of control neurons (diseased vs. control: OR=31.20, p = 2.35×10-10). Disease-associated sIndels were primarily two-basepair deletions resembling signature ID4, which was previously linked to topoisomerase 1 (TOP1)-mediated mutagenesis. Duplex sequencing confirmed the presence of sIndels and identified similar single-strand events as potential precursor lesions. TOP1-associated sIndel mutagenesis and resulting genome instability may thus represent a common mechanism of neurodegeneration.
Collapse
Affiliation(s)
- Zinan Zhou
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
| | | | - Guanlan Dong
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Bioinformatics and Integrative Genomics Program, Harvard Medical School; Boston, MA, USA
| | - Junho Kim
- Department of Biological Sciences, Sungkyunkwan University; Suwon, South Korea
| | - Jayoung Ku
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
| | - Kisong Kim
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
| | - Mingyun Bae
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
| | - Diane D. Shao
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Department of Neurology, Boston Children’s Hospital; Boston, MA, USA
| | - Bezawit Sahile
- Program in Neuroscience, Harvard Medical School; Boston, MA, USA
| | - Michael B. Miller
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Division of Neuropathology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - August Yue Huang
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - William J. Nathan
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Andre Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Peter J. Park
- Department of Biomedical Informatics, Harvard Medical School; Boston, MA, USA
| | - Clotilde Lagier-Tourenne
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School; Boston, MA, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Howard Hughes Medical Institute; Boston, MA, USA
| |
Collapse
|
48
|
Nie Y, Szebényi K, Wenger LMD, Lakatos A, Chinnery PF. Origin and cell type specificity of mitochondrial DNA mutations in C9ORF72 ALS-FTLD human brain organoids. SCIENCE ADVANCES 2025; 11:eadr0690. [PMID: 40053600 PMCID: PMC11887808 DOI: 10.1126/sciadv.adr0690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are primarily genetic in ~20% of patients. Mutations in C9ORF72 are the most frequent cause, but it is not understood why there is notable regional pathology. An increased burden of mitochondrial DNA (mtDNA) mutations in ALS-FTLD brains implicates mitochondrial mechanisms; however, it remains unclear how and when these mutations arise. To address this, we generated cerebral organoids derived from human-induced pluripotent stem cells (hiPSCs) of patients with ALS-FTLD harboring the C9ORF72 hexanucleotide repeat expansion alongside CRISPR-corrected isogenic and healthy controls. Here, we show a higher mtDNA single-nucleotide variant (mtSNV) burden in astroglia derived from C9ORF72-mutant organoids, with some de novo mtSNVs likely due to the C9ORF72 repeat and others evading selection to reach higher heteroplasmy levels. Thus, the functional consequences of the regional accumulation of mtSNVs in C9ORF72 ALS-FTLD brains are likely to manifest through astroglial mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yu Nie
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Kornélia Szebényi
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Lea M. D. Wenger
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - András Lakatos
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Patrick F. Chinnery
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
49
|
Vialle RA, de Paiva Lopes K, Li Y, Ng B, Schneider JA, Buchman AS, Wang Y, Farfel JM, Barnes LL, Wingo AP, Wingo TS, Seyfried NT, De Jager PL, Gaiteri C, Tasaki S, Bennett DA. Structural variants linked to Alzheimer's disease and other common age-related clinical and neuropathologic traits. Genome Med 2025; 17:20. [PMID: 40038788 PMCID: PMC11881306 DOI: 10.1186/s13073-025-01444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder with substantial genetic influence. While genome-wide association studies (GWAS) have identified numerous risk loci for late-onset AD (LOAD), the functional mechanisms underlying most of these associations remain unresolved. Large genomic rearrangements, known as structural variants (SVs), represent a promising avenue for elucidating such mechanisms within some of these loci. METHODS By leveraging data from two ongoing cohort studies of aging and dementia, the Religious Orders Study and Rush Memory and Aging Project (ROS/MAP), we performed genome-wide association analysis testing 20,205 common SVs from 1088 participants with whole genome sequencing (WGS) data. A range of Alzheimer's disease and other common age-related clinical and neuropathologic traits were examined. RESULTS First, we mapped SVs across 81 AD risk loci and discovered 22 SVs in linkage disequilibrium (LD) with GWAS lead variants and directly associated with the phenotypes tested. The strongest association was a deletion of an Alu element in the 3'UTR of the TMEM106B gene, in high LD with the respective AD GWAS locus and associated with multiple AD and AD-related disorders (ADRD) phenotypes, including tangles density, TDP-43, and cognitive resilience. The deletion of this element was also linked to lower TMEM106B protein abundance. We also found a 22-kb deletion associated with depression in ROS/MAP and bearing similar association patterns as GWAS SNPs at the IQCK locus. In addition, we leveraged our catalog of SV-GWAS to replicate and characterize independent findings in SV-based GWAS for AD and five other neurodegenerative diseases. Among these findings, we highlight the replication of genome-wide significant SVs for progressive supranuclear palsy (PSP), including markers for the 17q21.31 MAPT locus inversion and a 1483-bp deletion at the CYP2A13 locus, along with other suggestive associations, such as a 994-bp duplication in the LMNTD1 locus, suggestively linked to AD and a 3958-bp deletion at the DOCK5 locus linked to Lewy body disease (LBD) (P = 3.36 × 10-4). CONCLUSIONS While still limited in sample size, this study highlights the utility of including analysis of SVs for elucidating mechanisms underlying GWAS loci and provides a valuable resource for the characterization of the effects of SVs in neurodegenerative disease pathogenesis.
Collapse
Affiliation(s)
- Ricardo A Vialle
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA.
| | - Katia de Paiva Lopes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yan Li
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Jose M Farfel
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aliza P Wingo
- Department of Psychiatry, University of California, Davis, Davis, CA, USA
- VA Northern California Health Care System, Davis, CA, USA
| | - Thomas S Wingo
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Nicholas T Seyfried
- Department of Neurology and Department of Biochemistry, Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| |
Collapse
|
50
|
Wang HLV, Xiang JF, Yuan C, Veire AM, Gendron TF, Murray ME, Tansey MG, Hu J, Gearing M, Glass JD, Jin P, Corces VG, McEachin ZT. pTDP-43 levels correlate with cell type-specific molecular alterations in the prefrontal cortex of C9orf72 ALS/FTD patients. Proc Natl Acad Sci U S A 2025; 122:e2419818122. [PMID: 39999167 PMCID: PMC11892677 DOI: 10.1073/pnas.2419818122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Repeat expansions in the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis and familial frontotemporal dementia (ALS/FTD). To identify molecular defects that take place in the dorsolateral frontal cortex of patients with C9orf72 ALS/FTD, we compared healthy controls with C9orf72 ALS/FTD donor samples staged based on the levels of cortical phosphorylated TAR DNA binding protein (pTDP-43), a neuropathological hallmark of disease progression. We identified distinct molecular changes in different cell types that take place during FTD development. Loss of neurosurveillance microglia and activation of the complement cascade take place early, when pTDP-43 aggregates are absent or very low, and become more pronounced in late stages, suggesting an initial involvement of microglia in disease progression. Reduction of layer 2-3 cortical projection neurons with high expression of CUX2/LAMP5 also occurs early, and the reduction becomes more pronounced as pTDP-43 accumulates. Several unique features were observed only in samples with high levels of pTDP-43, including global alteration of chromatin accessibility in oligodendrocytes, microglia, and astrocytes; higher ratios of premature oligodendrocytes; increased levels of the noncoding RNA NEAT1 in astrocytes and neurons, and higher amount of phosphorylated ribosomal protein S6. Our findings reveal progressive functional changes in major cell types found in the prefrontal cortex of C9orf72 ALS/FTD patients that shed light on the mechanisms underlying the pathology of this disease.
Collapse
Affiliation(s)
- Hsiao-Lin V. Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA30322
| | - Jian-Feng Xiang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Chenyang Yuan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA30322
| | - Austin M. Veire
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL32224
| | | | | | - Malú G. Tansey
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL32607
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL32607
| | - Jian Hu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA30322
| | - Marla Gearing
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA30322
- Department of Neurology, Emory University School of Medicine, Atlanta, GA30322
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA30322
| | - Jonathan D. Glass
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA30322
- Department of Neurology, Emory University School of Medicine, Atlanta, GA30322
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA30322
| | - Victor G. Corces
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA30322
| | - Zachary T. McEachin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
- Emory Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA30322
| |
Collapse
|