1
|
Decker L, Menge S, Freischmidt A. Cryptic exon inclusion in TDP-43 proteinopathies: opportunities and challenges. Neural Regen Res 2025; 20:2003-2004. [PMID: 39254559 PMCID: PMC11691479 DOI: 10.4103/nrr.nrr-d-24-00459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Lorena Decker
- Department of Neurology, Ulm University, Ulm, Germany
| | - Sonja Menge
- Department of Neurology, Ulm University, Ulm, Germany
| | | |
Collapse
|
2
|
Balendra R, Sreedharan J, Hallegger M, Luisier R, Lashuel HA, Gregory JM, Patani R. Amyotrophic lateral sclerosis caused by TARDBP mutations: from genetics to TDP-43 proteinopathy. Lancet Neurol 2025; 24:456-470. [PMID: 40252666 DOI: 10.1016/s1474-4422(25)00109-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/07/2025] [Accepted: 03/20/2025] [Indexed: 04/21/2025]
Abstract
Mutations in the TARDBP gene, which encodes the TDP-43 protein, account for only 3-5% of familial cases of amyotrophic lateral sclerosis and less than 1% of cases that are apparently idiopathic. However, the discovery of neuronal inclusions of TDP-43 as the neuropathological hallmark in the majority of cases of amyotrophic lateral sclerosis has transformed our understanding of the pathomechanisms underlying neurodegeneration. An individual TARDBP mutation can cause phenotypic heterogeneity. Most mutations lie within the C-terminus of the TDP-43 protein. In pathological conditions, TDP-43 is mislocalised from the nucleus to the cytoplasm, where it can be phosphorylated, cleaved, and form insoluble aggregates. This mislocalisation leads to dysfunction of downstream pathways of RNA metabolism, proteostasis, mitochondrial function, oxidative stress, axonal transport, and local translation. Biomarkers for TDP-43 dysfunction and targeted therapies are being developed, justifying cautious optimism for personalised medicine approaches that could rescue the downstream effects of TDP-43 pathology.
Collapse
Affiliation(s)
- Rubika Balendra
- Human Stem Cells and Neurodegeneration Laboratory, The Francis Crick Institute, London, UK; UK Dementia Research Institute at UCL, London, UK.
| | - Jemeen Sreedharan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Martina Hallegger
- UK Dementia Research Institute at King's, London, UK; The Francis Crick Institute, London, UK; Oxford-GSK Institute of Molecular and Computational Medicine, Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Raphaëlle Luisier
- Genomics and Health Informatics Group, Idiap Research Institute, Martigny, Switzerland
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Qatar Foundation, Doha, Qatar
| | | | - Rickie Patani
- Human Stem Cells and Neurodegeneration Laboratory, The Francis Crick Institute, London, UK; Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
3
|
Simon C, Graves OK, Akeju O, McKay TB. Elevated TDP-43 serum levels associated with postoperative delirium following major cardiac surgery. Brain Behav Immun Health 2025; 45:100974. [PMID: 40134937 PMCID: PMC11932660 DOI: 10.1016/j.bbih.2025.100974] [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: 02/08/2025] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
Background Postoperative delirium is a recurring complication among vulnerable patients undergoing major cardiac surgery. While delirium has been associated with prodromal dementia, there is minimal evidence to support the causality of this nuanced relationship. Clarification as to how postoperative delirium might lead to neurodegenerative dementias, perhaps through evidence of contemporaneous biomarkers, would heighten the plausibility of a causal correlation. TAR DNA-binding protein 43 (TDP-43), a nuclear protein essential for transcriptional events, has been linked to pathological aggregation in Alzheimer's disease (AD) and AD-related dementias (ADRD). Methods Circulating TDP-43 levels in cardiac surgical patients aged 60 years and older were evaluated in a biobank derived from the Minimizing ICU Neurological Dysfunction with Dexmedetomidine-induced Sleep (MINDDS) clinical trial. Serum total TDP-43 levels, measured with a single molecule array (Simoa), were compared across preoperative and postoperative day one timepoints according to delirium status assessed using the Confusion Assessment Method (CAM). To investigate the temporal changes in serum TDP-43, an independent validation cohort of 25 patients aged 60 years and older undergoing major cardiac surgery was analyzed. Results Total serum TDP-43 levels increased by 16.5% (95% CI: 5.9%-27.9%, p = 0.0021) on postoperative day one compared to baseline levels. This increase was more pronounced in patients who experienced delirium (median increase of 55.1%, 95% CI: 22.9%-96.4%, p = 0.0002). Further, these findings were conserved in multiple logistic regression models adjusting for treatment, age, sex, and baseline cognitive scores. In the validation cohort, TDP-43 levels were found to be significantly elevated immediately following cardiopulmonary bypass from the baseline, with a gradual decrease by postoperative day one. Conclusions Our findings demonstrate that post-cardiac surgery delirium among vulnerable patients is associated with significant elevations in circulating TDP-43. This relationship suggests that TDP-43 may serve as a prognostic biomarker for acute neurological insults and blood-brain barrier integrity following cardiac surgery. Overall, our results provide mechanistic insights into the inter-relationship between postoperative delirium and subsequent cognitive impairment, potentially offering new avenues for early intervention in at-risk surgical patients.
Collapse
Affiliation(s)
- Christopher Simon
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Occam Kelly Graves
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Tina B. McKay
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
4
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [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: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
5
|
San Gil R, Walker AK. Unlocking Disease-Modifying Treatments for TDP-43-Mediated Neurodegeneration. Bioessays 2025; 47:e202400257. [PMID: 39901378 PMCID: PMC11931677 DOI: 10.1002/bies.202400257] [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: 10/31/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 02/05/2025]
Abstract
Neurons degenerate in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), causing progressive and inevitably fatal neurological decline. The best therapeutic strategies target underlying disease mediators, but after decades of intensive research, the causes of these neurodegenerative diseases remain elusive. Recently, coordinated activities of large consortia, increasing open access to large datasets, new methods such as cryo-transmission electron microscopy, and advancements in high-resolution omics technologies have offered new insights into the biology of disease that bring us closer to understanding mechanisms of neurodegeneration. In particular, improved understanding of the roles of the key pathological protein TAR DNA binding protein 43 (TDP-43) in disease has revealed intriguing new opportunities that provide hope for better diagnostic tools and effective treatments for ALS and FTD.
Collapse
Affiliation(s)
- Rebecca San Gil
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
- School of Medical SciencesFaculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Adam K. Walker
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
- Sydney Pharmacy SchoolFaculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
6
|
Peck A, Dadi A, Yavarow Z, Alfano LN, Anderson D, Arkin MR, Chou TF, D'Ambrosio ES, Diaz-Manera J, Dudley JP, Elder AG, Ghoshal N, Hart CE, Hart MM, Huryn DM, Johnson AE, Jones KB, Kimonis V, Kiskinis E, Lee EB, Lloyd TE, Mapstone M, Martin A, Meyer H, Mozaffar T, Onyike CU, Pfeffer G, Pindon A, Raman M, Richard I, Rubinsztein DC, Schiava M, Schütz AK, Shen PS, Southworth DR, Staffaroni AM, Taralio-Gravovac M, Weihl CC, Yao Q, Ye Y, Peck N. 2024 VCP International Conference: Exploring multi-disciplinary approaches from basic science of valosin containing protein, an AAA+ ATPase protein, to the therapeutic advancement for VCP-associated multisystem proteinopathy. Neurobiol Dis 2025; 207:106861. [PMID: 40037468 PMCID: PMC11960434 DOI: 10.1016/j.nbd.2025.106861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
Valosin-containing protein (VCP/p97) is a ubiquitously expressed AAA+ ATPase associated with numerous protein-protein interactions and critical cellular functions including protein degradation and clearance, mitochondrial homeostasis, DNA repair and replication, cell cycle regulation, endoplasmic reticulum-associated degradation, and lysosomal functions including autophagy and apoptosis. Autosomal-dominant missense mutations in the VCP gene may result in VCP-associated multisystem proteinopathy (VCP-MSP), a rare degenerative disorder linked to heterogeneous phenotypes including inclusion body myopathy (IBM) with Paget's disease of bone (PDB) and frontotemporal dementia (FTD) or IBMPFD, amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), parkinsonism, Charcot-Marie Tooth disease (CMT), and spastic paraplegia. The complexity of VCP-MSP makes collaboration among stakeholders essential and necessitates a multi-disciplinary approach. The 2024 VCP International Conference was hosted at Caltech between February 22 and 25. Co-organized by Cure VCP Disease and Dr. Tsui-Fen Chou, the meeting aimed to center the patient as a research partner, harmonize diverse stakeholder engagement, and bridge the gap between basic and clinical neuroscience as it relates to VCP-MSP. Over 100 multi-disciplinary experts attended, ranging from basic scientists to clinicians to patient advocates. Attendees discussed genetics and clinical presentation, cellular and molecular mechanisms underlying disease, therapeutic approaches, and strategies for future VCP research. The conference included three roundtable discussions, 29 scientific presentations, 32 scientific posters, nine patient and caregiver posters, and a closing discussion forum. The following conference proceedings summarize these sessions, highlighting both the identified gaps in knowledge and the significant strides made towards understanding and treating VCP diseases.
Collapse
Affiliation(s)
- A Peck
- Cure VCP Disease, Warner Robins, GA, USA
| | - A Dadi
- Cure VCP Disease, Warner Robins, GA, USA
| | - Z Yavarow
- Cure VCP Disease, Warner Robins, GA, USA
| | - L N Alfano
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | | | - M R Arkin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - T F Chou
- Department of Biology, Caltech, Pasadena, CA, USA
| | - E S D'Ambrosio
- Nationwide Children's Hospital, Columbus, OH, USA; Department of Genetic and Cellular Medicine and Department of Neurology, UMass Chan Medical School, Worcester, MA, USA
| | - J Diaz-Manera
- The John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, Center for Life, Central Parkway, Newcastle, Upon Tyne, United Kingdom
| | - J P Dudley
- LaMontagne Center for Infectious Disease, University of Texas, Austin, TX, USA
| | - A G Elder
- Cure VCP Disease, Warner Robins, GA, USA
| | - N Ghoshal
- Departments of Neurology and Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - C E Hart
- Creyon Bio, San Diego, CA, USA; Lilly, Indianapolis, IN, USA
| | - M M Hart
- Cure VCP Disease, Warner Robins, GA, USA
| | - D M Huryn
- Department of Chemistry University of Pennsylvania, Philadelphia, PA, USA
| | - A E Johnson
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - K B Jones
- Department of Orthopaedics, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - V Kimonis
- Department of Pediatrics, University of California, Irvine, CA, USA; Department of Neurology, University of California, Irvine, CA, USA
| | - E Kiskinis
- The Ken & Ruth Davee Department of Neurology, Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - E B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - T E Lloyd
- Department of Neurology Baylor College of Medicine, Houston, TX, USA
| | - M Mapstone
- Department of Neurology, University of California, Irvine, CA, USA
| | - A Martin
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - H Meyer
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - T Mozaffar
- Department of Neurology, University of California, Irvine, CA, USA
| | - C U Onyike
- Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - G Pfeffer
- Department of Clinical Neurosciences, University of Calgary, Calgary, Canada; Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - A Pindon
- Cure VCP Disease, Warner Robins, GA, USA; Myhre Syndrome Foundation, Richardson, TX, USA
| | - M Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - I Richard
- Généthon, 91000 Evry, France; Université Paris-Saclay, Université Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry, France
| | - D C Rubinsztein
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, UK; UK Dementia Research Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - M Schiava
- The John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, Center for Life, Central Parkway, Newcastle, Upon Tyne, United Kingdom
| | - A K Schütz
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - P S Shen
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - D R Southworth
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
| | - A M Staffaroni
- UCSF Memory and Aging Center University of California San Francisco, CA, USA
| | - M Taralio-Gravovac
- Department of Biochemistry & Molecular Biology, University of Calgary, Alberta, Calgary, Canada
| | - C C Weihl
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Q Yao
- Department of Medicine Stony Brook University, Renaissance School of Medicine, Stony Brook, NY, USA
| | - Y Ye
- Laboratory of Molecular Biology, NIH, NIDDK, Bethesda, MD, USA
| | - N Peck
- Cure VCP Disease, Warner Robins, GA, USA.
| |
Collapse
|
7
|
Ikenaga C, Wilson AB, Irwin KE, Peethambaran Mallika A, Kilgore C, Sinha IR, Michelle EH, Ling JP, Wong PC, Lloyd TE. Loss of TDP-43 Splicing Repression Occurs in Myonuclei of Inclusion Body Myositis Patients. Ann Neurol 2025; 97:629-641. [PMID: 39757935 DOI: 10.1002/ana.27167] [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: 05/05/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVE Inclusion body myositis (IBM) is an idiopathic inflammatory myopathy with muscle pathology characterized by endomysial inflammation, rimmed vacuoles, and cytoplasmic mislocalization of transactive response DNA-binding protein 43 (TDP-43). We aimed to determine whether loss of TDP-43 splicing repression led to the production of "cryptic peptides" that could be detected in muscle biopsies as a useful biomarker for IBM. METHODS We used an antisera against a neoepitope encoded by a TDP-43-dependent cryptic exon within hepatoma-derived growth factor-like protein 2 (HDGFL2) for immunohistochemical analysis on muscle biopsy samples of 122 patients with IBM, 181 disease controls, and 16 healthy controls without abnormal muscle pathology. In situ hybridization was also utilized to detect the localization of cryptic HDGFL2 transcripts. RESULTS We found cryptic HDGFL2 peptides localized within myonuclei from muscle biopsies in 79 of 122 patients with IBM (65%), and this staining correlated with TDP-43 depletion. In contrast, cryptic HDGFL2 immunoreactivity was absent in 197 muscle biopsies from a variety of disease controls, except for 2 patients with vacuolar myopathies. Notably, we show that cryptic HDGFL2 transcripts are accompanied by the detection of cryptic HDGFL2 in muscle fibers of IBM without rimmed vacuoles and TDP-43 aggregates. INTERPRETATION Together, our findings establish that loss of TDP-43 splicing repression occurs in myonuclei of IBM skeletal muscle and suggest that detection of cryptic peptides in muscle biopsies may be a useful biomarker. We suggest that a therapeutic strategy designed to restore TDP-43 function should be considered to attenuate the degeneration of skeletal muscle in this devastating disease. ANN NEUROL 2025;97:629-641.
Collapse
Affiliation(s)
- Chiseko Ikenaga
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew B Wilson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Katherine E Irwin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Collin Kilgore
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Irika R Sinha
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth H Michelle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jonathan P Ling
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Philip C Wong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thomas E Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Neurology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
8
|
Pickles SR, Gonzalez Bejarano J, Narayan A, Daughrity L, Maroto Cidfuentes C, Reeves MM, Yue M, Castellanos Otero P, Estades Ayuso V, Dunmore J, Song Y, Tong J, DeTure M, Rawlinson B, Castanedes‐Casey M, Dulski J, Cerquera‐Cleves C, Zhang Y, Josephs KA, Dickson DW, Petrucelli L, Wszolek ZK, Prudencio M. TDP-43 Cryptic RNAs in Perry Syndrome: Differences across Brain Regions and TDP-43 Proteinopathies. Mov Disord 2025; 40:662-671. [PMID: 39788898 PMCID: PMC12006891 DOI: 10.1002/mds.30104] [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: 10/10/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Perry syndrome (PS) is a rare and fatal hereditary autosomal dominant neurodegenerative disorder caused by mutations in dynactin (DCTN1). PS brains accumulate inclusions positive for ubiquitin, transactive-response DNA-binding protein of 43 kDa (TDP-43), and to a lesser extent dynactin. OBJECTIVES Little is known regarding the contributions of TDP-43, an RNA binding protein that represses cryptic exon inclusion, in PS. Therefore, we sought to identify the degree of TDP-43 dysfunction in two regions of PS brains. METHODS We evaluated the levels of insoluble pTDP-43 and TDP-43-regulated cryptic RNAs and protein in the caudate nucleus and substantia nigra of 7 PS cases, 12 cases of frontotemporal lobar degeneration (FTLD) with TDP-43 pathology, and 11 cognitively healthy controls without TDP-43 pathology. RESULTS Insoluble pTDP-43 protein levels were detected in PS brains to a similar extent in the caudate nucleus and substantia nigra but lower than those in FTLD brains. The caudate nucleus of PS showed accumulation of eight TDP-43-regulated cryptic RNAs (ACTL6B, CAMK2B, STMN2, UNC13A, KCNQ2, ATG4B, GPSM2, and HDGFL2) and cryptic protein (HDGFL2) characteristic of FTLD. Conversely, only one cryptic target, UNC13A, reached significance in the substantia nigra despite similar pTDP-43 levels. CONCLUSION We detected TDP-43 cryptic RNAs and protein in PS caudate nucleus. Given the importance of cryptic exon biology in the development of biomarkers, and the identification of novel targets for therapeutic intervention, it is imperative we understand the consequences of TDP-43 dysfunction across different brain regions and determine the targets that are specific and common to TDP-43 proteinopathies. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sarah R. Pickles
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | | | - Anand Narayan
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | | | - Mei Yue
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Virginia Estades Ayuso
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Judy Dunmore
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yuping Song
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Jimei Tong
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Michael DeTure
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Jaroslaw Dulski
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
- Division of Neurological and Psychiatric Nursing, Faculty of Health SciencesMedical University of GdanskGdanskPoland
- Neurology DepartmentSt Adalbert Hospital, Copernicus PL Ltd.GdanskPoland
| | - Catalina Cerquera‐Cleves
- Department of Neurosciences, Neurology UnitHospital Universitario San IgnacioBogotaColombia
- CHU de Québec Research Center, Axe NeurosciencesUniversité LavalQuebec CityQuebecCanada
| | - Yongjie Zhang
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | | | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Leonard Petrucelli
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | | | - Mercedes Prudencio
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| |
Collapse
|
9
|
Cao Y, Xu Y, Cao M, Chen N, Zeng Q, Lai MKP, Fan D, Sethi G, Cao Y. Fluid-based biomarkers for neurodegenerative diseases. Ageing Res Rev 2025; 108:102739. [PMID: 40122396 DOI: 10.1016/j.arr.2025.102739] [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: 09/01/2024] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Neurodegenerative diseases, such as Alzheimer's Disease (AD), Multiple Sclerosis (MS), Parkinson's Disease (PD), and Amyotrophic Lateral Sclerosis (ALS) are increasingly prevalent as global populations age. Fluid biomarkers, derived from cerebrospinal fluid (CSF), blood, saliva, urine, and exosomes, offer a promising solution for early diagnosis, prognosis, and disease monitoring. These biomarkers can reflect critical pathological processes like amyloid-beta (Aβ) deposition, tau protein hyperphosphorylation, α-syn misfolding, TDP-43 mislocalization and aggregation, and neuronal damage, enabling detection long before clinical symptoms emerge. Recent advances in blood-based biomarkers, particularly plasma Aβ, phosphorylated tau, and TDP-43, have shown diagnostic accuracy equivalent to CSF biomarkers, offering more accessible testing options. This review discusses the current challenges in fluid biomarker research, including variability, standardization, and sensitivity issues, and explores how combining multiple biomarkers with clinical symptoms improves diagnostic reliability. Ethical considerations, future directions involving extracellular vehicles (EVs), and the integration of artificial intelligence (AI) are also highlighted. Continued research efforts will be key to overcoming these obstacles, enabling fluid biomarkers to become crucial tools in personalized medicine for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Yifei Xu
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Nan Chen
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Qingling Zeng
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore; Memory, Aging and Cognition Centre, National University Health System, Singapore
| | - Dahua Fan
- Institute of Maternal-Fetal Medicine,Shunde Women and Children's Hospital, Guangdong Medical University, Foshan 528300, China.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China.
| |
Collapse
|
10
|
Thal DR, Poesen K, Vandenberghe R, De Meyer S. Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers. Mol Neurodegener 2025; 20:33. [PMID: 40087672 PMCID: PMC11907863 DOI: 10.1186/s13024-025-00819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.
Collapse
Affiliation(s)
- Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.
| | - Koen Poesen
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Steffi De Meyer
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Sheth U, Öijerstedt L, Heckman MG, White LJ, Heuer HW, Lario Lago A, Forsberg LK, Faber KM, Foroud TM, Rademakers R, Ramos EM, Appleby BS, Bozoki AC, Darby RR, Dickerson BC, Domoto-Reilly K, Galasko DR, Ghoshal N, Graff-Radford NR, Grant IM, Hales CM, Hsiung GYR, Huey ED, Irwin D, Kwan JY, Litvan I, Mackenzie IR, Masdeu JC, Mendez MF, Onyike CU, Pascual B, Pressman PS, Roberson ED, Snyder A, Tartaglia MC, Seeley WW, Dickson DW, Rosen HJ, Boeve BF, Boxer AL, Petrucelli L, Gendron TF. Comprehensive cross-sectional and longitudinal comparisons of plasma glial fibrillary acidic protein and neurofilament light across FTD spectrum disorders. Mol Neurodegener 2025; 20:30. [PMID: 40075459 PMCID: PMC11905702 DOI: 10.1186/s13024-025-00821-4] [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: 10/03/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Therapeutic development for frontotemporal dementia (FTD) is hindered by the lack of biomarkers that inform susceptibility/risk, prognosis, and the underlying causative pathology. Blood glial fibrillary acidic protein (GFAP) has garnered attention as a FTD biomarker. However, investigations of GFAP in FTD have been hampered by symptomatic and histopathologic heterogeneity and small cohort sizes contributing to inconsistent findings. Therefore, we evaluated plasma GFAP as a FTD biomarker and compared its performance to that of neurofilament light (NfL) protein, a leading FTD biomarker. METHODS We availed ARTFL LEFFTDS Longitudinal Frontotemporal Lobar Degeneration (ALLFTD) study resources to conduct a comprehensive cross-sectional and longitudinal examination of the susceptibility/risk, prognostic, and predictive performance of GFAP and NfL in the largest series of well-characterized presymptomatic FTD mutation carriers and participants with sporadic or familial FTD syndromes. Utilizing single molecule array technology, we measured GFAP and NfL in plasma from 161 controls, 127 presymptomatic mutation carriers, 702 participants with a FTD syndrome, and 67 participants with mild behavioral and/or cognitive changes. We used multivariable linear regression and Cox proportional hazard models adjusted for co-variates to examine the biomarker utility of baseline GFAP and NfL concentrations or their rates of change. RESULTS Compared to controls, GFAP and NfL were elevated in each FTD syndrome but GFAP, unlike NfL, poorly discriminated controls from participants with mild symptoms. Similarly, both baseline GFAP and NfL were higher in presymptomatic mutation carriers who later phenoconverted, but NfL better distinguished non-converters from phenoconverters. We additionally observed that GFAP and NfL were associated with disease severity indicators and survival, but NfL far outperformed GFAP. Nevertheless, we validated findings that the GFAP/NfL ratio may discriminate frontotemporal lobar degeneration with tau versus TDP-43 pathology. CONCLUSIONS Our head-to-head comparison of plasma GFAP and NfL as biomarkers for FTD indicate that NfL consistently outmatched GFAP as a prognostic and predictive biomarker for participants with a FTD syndrome, and as a susceptibility/risk biomarker for people at genetic risk of FTD. Our findings underscore the need to include leading biomarkers in investigations evaluating new biomarkers if the field is to fully ascertain their performance and clinical value.
Collapse
Affiliation(s)
- Udit Sheth
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Linn Öijerstedt
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Michael G Heckman
- Division of Clinical Trials and Biostatistics, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Launia J White
- Division of Clinical Trials and Biostatistics, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Hilary W Heuer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 91358, USA
| | - Argentina Lario Lago
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 91358, USA
| | - Leah K Forsberg
- Department of Neurology, Mayo Clinic, 200 First St, SW, Rochester, MN, 55905, USA
| | - Kelley M Faber
- Department of Medical and Molecular Genetics, The National Centralized Repository for Alzheimer's Disease and Related Dementias, 351 W. 10Th St TK-217, Indianapolis, IN, 46202, USA
| | - Tatiana M Foroud
- Department of Medical and Molecular Genetics, The National Centralized Repository for Alzheimer's Disease and Related Dementias, 351 W. 10Th St TK-217, Indianapolis, IN, 46202, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Eliana Marisa Ramos
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Reed Neurological Research Center, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Brian S Appleby
- Department of Neurology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Andrea C Bozoki
- Department of Neurology, University of North Carolina, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - R Ryan Darby
- Department of Neurology, Vanderbilt University, 1161 21St Ave S, Nashville, TN, 37212, USA
| | - Bradford C Dickerson
- Department of Neurology, Frontotemporal Disorders Unit, Massachusetts General Hospital and Harvard Medical School, 149 13th St, Boston, MA, 02129, USA
| | - Kimiko Domoto-Reilly
- Department of Neurology, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195-6465, USA
| | - Douglas R Galasko
- Department of Neurosciences, University of California, 9500 Gilman Drive, La Jolla, CA, 92037-0948, USA
| | - Nupur Ghoshal
- Departments of Neurology and Psychiatry, Washington University School of Medicine, Washington University, 660 South Euclid, St. Louis, MO, 63110, USA
| | - Neill R Graff-Radford
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Ian M Grant
- Department of Neurology, Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern Feinberg School of Medicine, 300 E. Superior, Tarry 8-715, Chicago, IL, 60610, USA
| | - Chadwick M Hales
- Center for Neurodegenerative Disease, Department of Neurology, Emory University School of Medicine and Emory, 12 Executive Park Drive, Atlanta, GA, 30329, USA
| | - Ging-Yuek Robin Hsiung
- Division of Neurology, University of British Columbia, S151-2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Edward D Huey
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, 345 Blackstone Boulevard, Providence, RI, 02906, USA
| | - David Irwin
- Department of Neurology and Penn Frontotemporal Degeneration Center, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce St, Philadelphia, PA, 19104, USA
| | - Justin Y Kwan
- Disorders and Stroke, National Institute of Neurological, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Irene Litvan
- Department of Neurosciences, University of California, 9452 Medical Center Drive, La Jolla, CA, 92037, USA
| | - Ian R Mackenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Joseph C Masdeu
- Department of Neurology, Houston Methodist Neurological Institute, Weill Cornell Medicine, 6560 Fannin St, Houston, TX, 77030, USA
| | - Mario F Mendez
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Reed Neurological Research Center, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Chiadi U Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Belen Pascual
- Department of Neurology, Houston Methodist Neurological Institute, Weill Cornell Medicine, 6560 Fannin St, Houston, TX, 77030, USA
| | - Peter S Pressman
- Department of Neurology, University of Colorado School of Medicine, 12631 East 17Th Avenue, Aurora, CO, 80045, USA
- Layton Aging and Alzheimer's Disease Research Center, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Erik D Roberson
- Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35233, USA
| | - Allison Snyder
- Disorders and Stroke, National Institute of Neurological, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - M Carmela Tartaglia
- Division of Neurology, Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 6 Queen's Park Crescent West, Third Floor, Toronto, ON, M5S 3H2, Canada
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 91358, USA
- Department of Pathology, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 91358, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, 200 First St, SW, Rochester, MN, 55905, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 91358, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
12
|
Dang M, Wu L, Zhang X. Structural insights and milestones in TDP-43 research: A comprehensive review of its pathological and therapeutic advances. Int J Biol Macromol 2025; 306:141677. [PMID: 40032118 DOI: 10.1016/j.ijbiomac.2025.141677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Transactive response (TAR) DNA-binding protein 43 (TDP-43) is a critical RNA/DNA-binding protein involved in various cellular processes, including RNA splicing, transcription regulation, and RNA stability. Mislocalization and aggregation of TDP-43 in the cytoplasm are key features of the pathogenesis of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Alzheimer's disease (AD). This review provides a comprehensive retrospective and prospective analysis of TDP-43 research, highlighting structural insights, significant milestones, and the evolving understanding of its physiological and pathological functions. We delineate five major stages in TDP-43 research, from its initial discovery as a pathological hallmark in neurodegeneration to the recent advances in understanding its liquid-liquid phase separation (LLPS) behavior and interactions with cellular processes. Furthermore, we assess therapeutic strategies targeting TDP-43 pathology, categorizing approaches into direct and indirect interventions, alongside modulating aberrant TDP-43 LLPS. We propose that future research will focus on three critical areas: targeting TDP-43 structural polymorphisms for disease-specific therapeutics, exploring dual temporal-spatial modulation of TDP-43, and advancing nano-therapy. More importantly, we emphasize the importance of understanding TDP-43's functional repertoire at the mesoscale, which bridges its molecular functions with broader cellular processes. This review offers a foundational framework for advancing TDP-43 research and therapeutic development.
Collapse
Affiliation(s)
- Mei Dang
- Chinese-German Joint Institute for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China; Department of Biological Sciences, Faculty of Science, National University of Singapore, 10 Kent Ridge Crescent, 119260, Singapore
| | - Longjiang Wu
- Chinese-German Joint Institute for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China
| | - Xiaoying Zhang
- Chinese-German Joint Institute for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China; Centre of Molecular & Environmental Biology, Department of Biology, University of Minho, 4710-057 Braga, Portugal; Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, N1G 2W1 Guelph, Ontario, Canada.
| |
Collapse
|
13
|
Wischnewski S, Rausch HW, Ikenaga C, Leipe J, Lloyd TE, Schirmer L. Emerging mechanisms and therapeutics in inflammatory muscle diseases. Trends Pharmacol Sci 2025; 46:249-263. [PMID: 39939222 DOI: 10.1016/j.tips.2025.01.005] [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: 11/15/2024] [Revised: 01/03/2025] [Accepted: 01/15/2025] [Indexed: 02/14/2025]
Abstract
Idiopathic inflammatory myopathies (IIMs), or myositis, are rare diseases marked by immune-driven muscle damage and complications like skin lesions and interstitial lung disease (ILD). Despite advances, challenges in diagnosis and treatment persist, particularly in inclusion body myositis (IBM), where no effective therapy exists. Recent breakthroughs, including transcriptomics and insights into antibody-mediated immunity and interferon (IFN) signaling, have clarified IIM pathophysiology and spurred the development of new therapies, such as chimeric antigen receptor (CAR) T cells and Janus kinase (JAK) inhibitors. We explore the latest findings on the mechanisms underlying adult-onset IIMs, emphasizing IBM pathobiology and its unique immune and degenerative pathways, such as a selective type 2 myofiber damage and severe cell stress. Finally, we highlight the recent advances in transcriptomics, single-cell analysis, and machine learning in transforming IIM research by improving diagnostic accuracy, uncovering therapeutic targets, and supporting the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Sven Wischnewski
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hans-Werner Rausch
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Chiseko Ikenaga
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Kitasato University Hospital, Tokyo, Japan
| | - Jan Leipe
- Division of Rheumatology, Department of Medicine V, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Division of Rheumatology, Department of Internal Medicine I, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - Thomas E Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Baylor College of Medicine, Houston, TX, USA.
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
14
|
Mizielinska S, Hautbergue GM, Gendron TF, van Blitterswijk M, Hardiman O, Ravits J, Isaacs AM, Rademakers R. Amyotrophic lateral sclerosis caused by hexanucleotide repeat expansions in C9orf72: from genetics to therapeutics. Lancet Neurol 2025; 24:261-274. [PMID: 39986312 PMCID: PMC12010636 DOI: 10.1016/s1474-4422(25)00026-2] [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: 08/05/2024] [Revised: 11/22/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025]
Abstract
GGGGCC repeat expansions in C9orf72 are a common genetic cause of amyotrophic lateral sclerosis in people of European ancestry; however, substantial variability in the penetrance of the mutation, age at disease onset, and clinical presentation can complicate diagnosis and prognosis. The repeat expansion is bidirectionally transcribed in the sense and antisense directions into repetitive RNAs and translated into dipeptide repeat proteins, and both accumulate in the cortex, cerebellum, and the spinal cord. Furthermore, neuropathological aggregates of phosphorylated TDP-43 are observed in motor cortex and other cortical regions, and in the spinal cord of patients at autopsy. C9orf72 repeat expansions can also cause frontotemporal dementia. The GGGGCC repeat induces a complex interplay of loss-of-function and gain-of-function pathological mechanisms. Clinical trials using antisense oligonucleotides to target the GGGGCC repeat RNA have not been successful, potentially because they only target a single gain-of-function mechanism. Novel therapeutic approaches targeting the DNA repeat expansion, multiple repeat-derived RNA species, or downstream targets of TDP-43 dysfunction are, however, on the horizon, together with the development of diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Sarah Mizielinska
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN), Neuroscience Institute, and Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, UK
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John Ravits
- Department of Neurosciences, ALS Translational Research, University of California San Diego, La Jolla, CA, USA
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| | - 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
|
15
|
Mamede LD, Hu M, Titus AR, Vaquer-Alicea J, French RL, Diamond MI, Miller TM, Ayala YM. TDP-43 Aggregate Seeding Impairs Autoregulation and Causes TDP-43 Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637743. [PMID: 39990366 PMCID: PMC11844547 DOI: 10.1101/2025.02.11.637743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The aggregation, cellular mislocalization and dysfunction of TDP-43 are hallmarks of multiple neurodegenerative disorders. We find that inducing TDP-43 aggregation through prion-like seeding gradually diminishes normal TDP-43 nuclear localization and function. Aggregate-affected cells show signature features of TDP-43 loss of function, such as DNA damage and dysregulated TDP-43-target expression. We also observe strong activation of TDP-43-controlled cryptic exons in cells, including human neurons treated with proteopathic seeds. Furthermore, aggregate seeding impairs TDP-43 autoregulation, an essential mechanism controlling TDP-43 homeostasis. Interestingly, proteins that normally interact with TDP-43 are not recruited to aggregates, while other factors linked to TDP-43 pathology, including Ataxin 2, specifically colocalize to inclusions and modify seeding-induced aggregation. Our findings indicate that TDP-43 aggregation, mislocalization and loss of function are strongly linked and suggest that disruption of TDP-43 autoregulation establishes a toxic feed-forward mechanism that amplifies aggregation and may be central in mediating this pathological connection.
Collapse
Affiliation(s)
- Lohany Dias Mamede
- Edward Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis MO 63104, USA
| | - Miwei Hu
- Department of Neurology, Washington University in St. Louis, St. Louis MO 63110, USA
| | - Amanda R Titus
- Edward Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis MO 63104, USA
| | - Jaime Vaquer-Alicea
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Timothy M Miller
- Department of Neurology, Washington University in St. Louis, St. Louis MO 63110, USA
| | - Yuna M Ayala
- Edward Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis MO 63104, USA
| |
Collapse
|
16
|
Nguyen TB, Miramontes R, Chillon-Marinas C, Maimon R, Vazquez-Sanchez S, Lau AL, McClure NR, Wu Z, Wang KQ, England WE, Singha M, Stocksdale JT, Heath M, Jang KH, Jung S, Ling K, Jafar-Nejad P, McKnight JI, Ho LN, Dalahmah OA, Faull RLM, Steffan JS, Reidling JC, Jang C, Lee G, Cleveland DW, Lagier-Tourenne C, Spitale RC, Thompson LM. Aberrant splicing in Huntington's disease accompanies disrupted TDP-43 activity and altered m6A RNA modification. Nat Neurosci 2025; 28:280-292. [PMID: 39762660 PMCID: PMC11802453 DOI: 10.1038/s41593-024-01850-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 11/14/2024] [Indexed: 01/15/2025]
Abstract
Huntington's disease (HD) is caused by a CAG repeat expansion in the HTT gene, leading to altered gene expression. However, the mechanisms leading to disrupted RNA processing in HD remain unclear. Here we identify TDP-43 and the N6-methyladenosine (m6A) writer protein METTL3 to be upstream regulators of exon skipping in multiple HD systems. Disrupted nuclear localization of TDP-43 and cytoplasmic accumulation of phosphorylated TDP-43 occurs in HD mouse and human brains, with TDP-43 also co-localizing with HTT nuclear aggregate-like bodies distinct from mutant HTT inclusions. The binding of TDP-43 onto RNAs encoding HD-associated differentially expressed and aberrantly spliced genes is decreased. Finally, m6A RNA modification is reduced on RNAs abnormally expressed in the striatum of HD R6/2 mouse brain, including at clustered sites adjacent to TDP-43 binding sites. Our evidence supports TDP-43 loss of function coupled with altered m6A modification as a mechanism underlying alternative splicing in HD.
Collapse
Affiliation(s)
- Thai B Nguyen
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | | | - Carlos Chillon-Marinas
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Roy Maimon
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Sonia Vazquez-Sanchez
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alice L Lau
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Nicolette R McClure
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Zhuoxing Wu
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Keona Q Wang
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Whitney E England
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Monika Singha
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Jennifer T Stocksdale
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Marie Heath
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Ki-Hong Jang
- Department of Microbiology and Molecular Genetics, Chao Family Comprehensive Cancer Center, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Karen Ling
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | | | - Jharrayne I McKnight
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Leanne N Ho
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Osama Al Dalahmah
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
| | - Joan S Steffan
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, USA
| | | | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Gina Lee
- Department of Microbiology and Molecular Genetics, Chao Family Comprehensive Cancer Center, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Clotilde Lagier-Tourenne
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA.
| | - Leslie M Thompson
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA.
- UCI MIND, University of California, Irvine, Irvine, CA, USA.
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, USA.
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
17
|
Faller KME, Chaytow H, Gillingwater TH. Targeting common disease pathomechanisms to treat amyotrophic lateral sclerosis. Nat Rev Neurol 2025; 21:86-102. [PMID: 39743546 DOI: 10.1038/s41582-024-01049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
The motor neuron disease amyotrophic lateral sclerosis (ALS) is a devastating condition with limited treatment options. The past few years have witnessed a ramping up of translational ALS research, offering the prospect of disease-modifying therapies. Although breakthroughs using gene-targeted approaches have shown potential to treat patients with specific disease-causing mutations, the applicability of such therapies remains restricted to a minority of individuals. Therapies targeting more general mechanisms that underlie motor neuron pathology in ALS are therefore of considerable interest. ALS pathology is associated with disruption to a complex array of key cellular pathways, including RNA processing, proteostasis, metabolism and inflammation. This Review details attempts to restore cellular homeostasis by targeting these pathways in order to develop effective, broadly-applicable ALS therapeutics.
Collapse
Affiliation(s)
- Kiterie M E Faller
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Helena Chaytow
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
18
|
Novel Biomarkers of FTD-ALS. Neuroscientist 2025; 31:6. [PMID: 39927436 DOI: 10.1177/10738584241308752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
|
19
|
Li Y, Sun S. RNA dysregulation in neurodegenerative diseases. EMBO J 2025; 44:613-638. [PMID: 39789319 PMCID: PMC11790913 DOI: 10.1038/s44318-024-00352-6] [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: 06/19/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Dysregulation of RNA processing has in recent years emerged as a significant contributor to neurodegeneration. The diverse mechanisms and molecular functions underlying RNA processing underscore the essential role of RNA regulation in maintaining neuronal health and function. RNA molecules are bound by RNA-binding proteins (RBPs), and interactions between RNAs and RBPs are commonly affected in neurodegeneration. In this review, we highlight recent progress in understanding dysregulated RNA-processing pathways and the causes of RBP dysfunction across various neurodegenerative diseases. We discuss both established and emerging mechanisms of RNA-mediated neuropathogenesis in this rapidly evolving field. Furthermore, we explore the development of potential RNA-targeting therapeutic approaches for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- 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
| | - 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.
- Departments of Neuroscience, Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
20
|
Jellinger KA. The spectrum of behavioral disorders in amyotrophic lateral sclerosis: current view. J Neural Transm (Vienna) 2025; 132:217-236. [PMID: 39402174 DOI: 10.1007/s00702-024-02841-8] [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: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 02/02/2025]
Abstract
Behavioral disorders, with an average prevalence of 30-60% are important non-motor symptoms in amyotrophic lateral sclerosis (ALS) that have a negative impact on prognosis, management and quality of life, yet the underlying neurobiology is poorly understood. Among people with ALS, apathy, fatigue, anxiety, irritability and other behavioral symptoms are the most prominent, although less frequent than cognitive impairment. The present review explores the current understanding of behavioral changes in ALS with particular emphasis on our current knowledge about their structural and functional brain correlates, substantiating a multisystem degeneration with particular dysfunction of frontal-subcortical circuits and dysfunction of fronto-striatal, frontotemporal and other essential brain systems. The natural history of behavioral dysfunctions in ALS and their relationship to frontotemporal lobe degeneration (FTLD) are not fully understood, although they form a clinical continuum, suggesting a differential vulnerability of non-motor brain networks, ALS being considered a brain network disorder. An assessment of risks or the early detection of brain connectivity signatures before structural changes may be helpful in investigating the pathophysiological mechanisms of behavioral impairment in ALS. Treatment of both ALS and co-morbid behavioral disorders is a multidisciplinary task, but whereas no causal or disease-modifying therapies for ALS are available, symptomatic treatment of a variety of behavioral symptoms plays a pivotal role in patient care, although the management of behavioral symptoms in clinical care still remains limited.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, Vienna, A-1150, Austria.
| |
Collapse
|
21
|
Kempthorne L, Vaizoglu D, Cammack AJ, Carcolé M, Roberts MJ, Mikheenko A, Fisher A, Suklai P, Muralidharan B, Kroll F, Moens TG, Yshii L, Verschoren S, Hölbling BV, Moreira FC, Katona E, Coneys R, de Oliveira P, Zhang YJ, Jansen K, Daughrity LM, McGown A, Ramesh TM, Van Den Bosch L, Lignani G, Rahim AA, Coyne AN, Petrucelli L, Rihel J, Isaacs AM. Dual-targeting CRISPR-CasRx reduces C9orf72 ALS/FTD sense and antisense repeat RNAs in vitro and in vivo. Nat Commun 2025; 16:459. [PMID: 39779704 PMCID: PMC11711508 DOI: 10.1038/s41467-024-55550-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: 01/23/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The most common genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) is an intronic G4C2 repeat expansion in C9orf72. The repeats undergo bidirectional transcription to produce sense and antisense repeat RNA species, which are translated into dipeptide repeat proteins (DPRs). As toxicity has been associated with both sense and antisense repeat-derived RNA and DPRs, targeting both strands may provide the most effective therapeutic strategy. CRISPR-Cas13 systems mature their own guide arrays, allowing targeting of multiple RNA species from a single construct. We show CRISPR-Cas13d variant CasRx effectively reduces overexpressed C9orf72 sense and antisense repeat transcripts and DPRs in HEK cells. In C9orf72 patient-derived iPSC-neuron lines, CRISPR-CasRx reduces endogenous sense and antisense repeat RNAs and DPRs and protects against glutamate-induced excitotoxicity. AAV delivery of CRISPR-CasRx to two distinct C9orf72 repeat mouse models significantly reduced both sense and antisense repeat-containing transcripts. This highlights the potential of RNA-targeting CRISPR systems as therapeutics for C9orf72 ALS/FTD.
Collapse
Affiliation(s)
- Liam Kempthorne
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Deniz Vaizoglu
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alexander J Cammack
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Martha J Roberts
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alla Mikheenko
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alessia Fisher
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Pacharaporn Suklai
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Bhavana Muralidharan
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, 560065, India
| | - François Kroll
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Thomas G Moens
- VIB-KU Center for Brain and Disease Research, Leuven, 3001, Belgium
| | - Lidia Yshii
- VIB-KU Center for Brain and Disease Research, Leuven, 3001, Belgium
| | - Stijn Verschoren
- VIB-KU Center for Brain and Disease Research, Leuven, 3001, Belgium
| | - Benedikt V Hölbling
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Francisco C Moreira
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Eszter Katona
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Rachel Coneys
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Paula de Oliveira
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Karen Jansen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Alexander McGown
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Tennore M Ramesh
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | | | - Gabriele Lignani
- Department of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alyssa N Coyne
- Department of Neurology, Johns Hopkins University, Baltimore, USA
- Brain Science Institute, Johns Hopkins University, Baltimore, USA
| | | | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
| |
Collapse
|
22
|
Kõks S, Rallmann K, Muldmaa M, Price J, Pfaff AL, Taba P. Whole blood transcriptome profile identifies motor neurone disease RNA biomarker signatures. Exp Biol Med (Maywood) 2025; 249:10401. [PMID: 39844875 PMCID: PMC11750576 DOI: 10.3389/ebm.2024.10401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Blood-based biomarkers for motor neuron disease are needed for better diagnosis, progression prediction, and clinical trial monitoring. We used whole blood-derived total RNA and performed whole transcriptome analysis to compare the gene expression profiles in (motor neurone disease) MND patients to the control subjects. We compared 42 MND patients to 42 aged and sex-matched healthy controls and described the whole transcriptome profile characteristic for MND. In addition to the formal differential analysis, we performed functional annotation of the genomics data and identified the molecular pathways that are differentially regulated in MND patients. We identified 12,972 genes differentially expressed in the blood of MND patients compared to age and sex-matched controls. Functional genomic annotation identified activation of the pathways related to neurodegeneration, RNA transcription, RNA splicing and extracellular matrix reorganisation. Blood-based whole transcriptomic analysis can reliably differentiate MND patients from controls and can provide useful information for the clinical management of the disease and clinical trials.
Collapse
Affiliation(s)
- Sulev Kõks
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Karin Rallmann
- Department of Neurology, Tartu University Hospital, Tartu, Estonia
| | - Mari Muldmaa
- Department of Neurology, North Estonia Medical Center, Tallinn, Estonia
- Institute of Clinical Medicine, University Tartu, Tartu, Estonia
| | - Jack Price
- Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Abigail L. Pfaff
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Pille Taba
- Institute of Clinical Medicine, University Tartu, Tartu, Estonia
| |
Collapse
|
23
|
Benatar M, Robertson J, Andersen PM. Amyotrophic lateral sclerosis caused by SOD1 variants: from genetic discovery to disease prevention. Lancet Neurol 2025; 24:77-86. [PMID: 39706636 DOI: 10.1016/s1474-4422(24)00479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/19/2024] [Accepted: 11/15/2024] [Indexed: 12/23/2024]
Abstract
Pathogenic variants in the superoxide dismutase 1 (SOD1) gene were the first identified genetic cause of amyotrophic lateral sclerosis (ALS), in 1993. This discovery enabled the development of transgenic rodent models for studying the biology of SOD1 ALS. The understanding that SOD1 ALS is driven by a toxic gain-of-function mutation has led to therapeutic strategies that aim to lower concentrations of SOD1 protein, an endeavour that has been complicated by the phenotypic heterogeneity of SOD1 ALS. The successful development of genetically targeted therapies to reduce SOD1 expression, together with a better understanding of pre-symptomatic disease and the discovery of neurofilament light protein as a susceptibility/risk biomarker that predicts phenoconversion, has ushered in a new era of trials that aim to prevent clinically manifest SOD1 ALS. The 30-year journey from gene discovery to gene therapy has not only uncovered the pathophysiology of SOD1 ALS, but has also facilitated the development of biomarkers that should aid therapy development for all forms of ALS.
Collapse
Affiliation(s)
- Michael Benatar
- Department of Neurology and ALS Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Janice Robertson
- University of Toronto, Tanz Centre for Research in Neurodegenerative Diseases, Department of Laboratory Medicine and Pathobiology, Toronto, ON, Canada
| | | |
Collapse
|
24
|
Rothstein JD, Hoke A, Mohassel P. Advances in neuromuscular disorders 2024. Lancet Neurol 2025; 24:18-20. [PMID: 39706622 DOI: 10.1016/s1474-4422(24)00489-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Affiliation(s)
- Jeffrey D Rothstein
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Payam Mohassel
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
25
|
Keeley O, Mendoza E, Menon D, Coyne AN. CHMP2B promotes CHMP7 mediated nuclear pore complex injury in sporadic ALS. Acta Neuropathol Commun 2024; 12:199. [PMID: 39709457 PMCID: PMC11662732 DOI: 10.1186/s40478-024-01916-7] [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: 11/11/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Abstract
Alterations to the composition and function of neuronal nuclear pore complexes (NPCs) have been documented in multiple neurodegenerative diseases including Amyotrophic Lateral Sclerosis (ALS). Moreover, recent work has suggested that injury to the NPC can at least in part contribute to TDP-43 loss of function and mislocalization, a pathological hallmark of ALS and related neurodegenerative diseases. Collectively, these studies highlight a role for disruptions in NPC homeostasis and surveillance as a significant pathophysiologic event in neurodegeneration. The ESCRT-III nuclear surveillance pathway plays a critical role in the surveillance and maintenance of NPCs and the surrounding nuclear environment. Importantly, pathologic alterations to this pathway and its protein constituents have been implicated in neurodegenerative diseases such as ALS. However, the mechanism by which this pathway contributes to disease associated alterations in the NPC remains unknown. Here we use an induced pluripotent stem cell (iPSC) derived neuron (iPSN) model of sALS to demonstrate that CHMP7/ESCRT-III nuclear maintenance/surveillance is overactivated in sALS neurons. This overactivation is dependent upon the ESCRT-III protein CHMP2B and sustained CHMP2B dependent "activation" is sufficient to contribute to pathologic CHMP7 nuclear accumulation and POM121 reduction. Importantly, partial knockdown of CHMP2B was sufficient to alleviate NPC injury and downstream TDP-43 dysfunction in sALS neurons thereby highlighting CHMP2B as a potential therapeutic target in disease.
Collapse
Affiliation(s)
- Olivia Keeley
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Emma Mendoza
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Druv Menon
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Alyssa N Coyne
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
26
|
Ito D, Okada K. Rethinking antisense oligonucleotide therapeutics for amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2024; 11:3054-3063. [PMID: 39473221 DOI: 10.1002/acn3.52234] [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: 09/16/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/18/2024] Open
Abstract
Antisense oligonucleotides, which are used to silence target genes, are gaining attention as a novel drug discovery modality for proteinopathies. However, while clinical trials for neurodegenerative diseases like amyotrophic lateral sclerosis have been conducted in recent years, the results have not always been favorable. The results from a Phase III trial of the antisense oligonucleotide, that is, tofersen, which targets SOD1 mRNA, showed decreased levels of cerebrospinal fluid SOD1 and plasma neurofilament light chain but no improvements in primary clinical endpoint. Moreover, case reports pertaining to patients with amyotrophic lateral sclerosis carrying FUS and C9orf72 mutations who received antisense oligonucleotide-based treatments have demonstrated a notable reduction in the targeted protein (thus providing the proof of mechanism) but with no discernible clinical benefits. There are several possible reasons why antisense oligonucleotides knockdown fails to achieve proof of concept, which need to be addressed: on-target adverse effects resulting from the loss of function of target gene and irreversible neuronal death cascade due to toxic protein accumulation, among other factors. This review provides an overview of the current status and discusses the prospects of antisense oligonucleotides treatment for amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Daisuke Ito
- Memory Center, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kensuke Okada
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Liampas I, Kyriakoulopoulou P, Karakoida V, Kavvoura PA, Sgantzos M, Bogdanos DP, Stamati P, Dardiotis E, Siokas V. Blood-Based Biomarkers in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:11838. [PMID: 39519389 PMCID: PMC11546606 DOI: 10.3390/ijms252111838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
This narrative review explores the current landscape of blood biomarkers in Frontotemporal dementia (FTD). Neurofilament light chain (NfL) may be useful in the differentiation of behavioral variant FTD from primary psychiatric disorders (PPDs) or dementia with Lewy bodies (DLB). In prodromal FTD and presymptomatic mutation carriers (GRN, MAPT, C9orf72), elevated NfL may herald pheno-conversion to full-blown dementia. Baseline NfL correlates with steeper neuroanatomical changes and cognitive, behavioral and functional decline, making NfL promising in monitoring disease progression. Phosphorylated neurofilament heavy chain (pNfH) levels have a potential limited role in the demarcation of the conversion stage to full-blown FTD. Combined NfL and pNfH measurements may allow a wider stage stratification. Total tau levels lack applicability in the framework of FTD. p-tau, on the other hand, is of potential value in the discrimination of FTD from Alzheimer's dementia. Progranulin concentrations could serve the identification of GRN mutation carriers. Glial fibrillary acidic protein (GFAP) may assist in the differentiation of PPDs from behavioral variant FTD and the detection of GRN mutation carriers (additional research is warranted). Finally, TAR DNA-binding protein-43 (TDP-43) appears to be a promising diagnostic biomarker for FTD. Its potential in distinguishing TDP-43 pathology from other FTD-related pathologies requires further research.
Collapse
Affiliation(s)
- Ioannis Liampas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| | | | - Vasiliki Karakoida
- School of Medicine, University of Patras, 26504 Rio Patras, Greece; (P.K.); (V.K.); (P.A.K.)
| | | | - Markos Sgantzos
- Department of Anatomy, Medical School, University of Thessaly, 41100 Larissa, Greece;
| | - Dimitrios P. Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece;
| | - Polyxeni Stamati
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (P.S.); (E.D.); (V.S.)
| |
Collapse
|
28
|
Bampton A, McHutchison C, Talbot K, Benatar M, Thompson AG, Turner MR. The Basis of Cognitive and Behavioral Dysfunction in Amyotrophic Lateral Sclerosis. Brain Behav 2024; 14:e70115. [PMID: 39501538 PMCID: PMC11538089 DOI: 10.1002/brb3.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 11/09/2024] Open
Abstract
OBJECTIVE To summarize and evaluate evidence pertaining to the clinical, genetic, histopathological, and neuroimaging correlates of cognitive and behavioral dysfunction in amyotrophic lateral sclerosis (ALS). METHODOLOGY We comprehensively reviewed the literature on cognitive and behavioral manifestations of ALS, narrating findings from both cross-sectional and longitudinal studies. We discussed knowledge gaps in the evidence base and key limitations affecting studies to date, before formulating a framework for future research paradigms aimed at investigating clinicopathological correlates of neuropsychological dysfunction in ALS. RESULTS Studies have demonstrated clinical associations with cognitive dysfunction in ALS e.g., bulbar-onset of symptoms, pathological associations (extramotor TDP-43 deposition), and imaging associations (frontotemporal involvement). The most common behavioral deficit, apathy, is highly associated with verbal fluency, but longitudinal studies assessing behavioral dysfunction in ALS are comparatively lacking. CONCLUSION Longitudinal studies have been helpful in identifying several potential correlates of cognitive and behavioral dysfunction but have frequently been confounded by selection bias and inappropriate testing platforms. This review provides a framework for more robust assessment of clinicopathological associations of neuropsychological abnormalities in ALS in the future, advocating for greater utilization of pre-symptomatic C9orf72 repeat expansion-carrying cohorts.
Collapse
Affiliation(s)
- Alexander Bampton
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | | | - Kevin Talbot
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Michael Benatar
- Department of NeurologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | | | - Martin R. Turner
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| |
Collapse
|
29
|
Zeng Y, Gitler AD. Regulators, mount up. Science 2024; 386:24-25. [PMID: 39361768 DOI: 10.1126/science.ads5951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Cryptic exons enable delivery of therapies only to sick neurons in a motor neuron disease.
Collapse
Affiliation(s)
- Yi Zeng
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
30
|
Kaul M, Mukherjee D, Weiner HL, Cox LM. Gut microbiota immune cross-talk in amyotrophic lateral sclerosis. Neurotherapeutics 2024; 21:e00469. [PMID: 39510899 PMCID: PMC11585889 DOI: 10.1016/j.neurot.2024.e00469] [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: 09/18/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by the loss of motor neurons. While there has been significant progress in defining the genetic contributions to ALS, greater than 90 % of cases are sporadic, which suggests an environmental component. The gut microbiota is altered in ALS and is an ecological factor that contributes to disease by modulating immunologic, metabolic, and neuronal signaling. Depleting the microbiome worsens disease in the SOD1 ALS animal model, while it ameliorates disease in the C9orf72 model of ALS, indicating critical subtype-specific interactions. Furthermore, administering beneficial microbiota or microbial metabolites can slow disease progression in animal models. This review discusses the current state of microbiome research in ALS, including interactions with different ALS subtypes, evidence in animal models and human studies, key immunologic and metabolomic mediators, and a path toward microbiome-based therapies for ALS.
Collapse
Affiliation(s)
- Megha Kaul
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, 02115, USA
| | - Debanjan Mukherjee
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, 02115, USA.
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Dargan R, Mikheenko A, Johnson NL, Packer B, Li Z, Craig EJ, Sarbanes SL, Bereda C, Mehta PR, Keuss M, Nalls MA, Qi YA, Weller CA, Fratta P, Ryan VH. Altered mRNA transport and local translation in iNeurons with RNA binding protein knockdown. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615153. [PMID: 39386562 PMCID: PMC11463369 DOI: 10.1101/2024.09.26.615153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Neurons rely on mRNA transport and local translation to facilitate rapid protein synthesis in processes far from the cell body. These processes allow precise spatial and temporal control of translation and are mediated by RNA binding proteins (RBPs), including those known to be associated with neurodegenerative diseases. Here, we use proteomics, transcriptomics, and microscopy to investigate the impact of RBP knockdown on mRNA transport and local translation in iPSC-derived neurons. We find thousands of transcripts enriched in neurites and that many of these transcripts are locally translated, possibly due to the shorter length of transcripts in neurites. Loss of frontotemporal dementia/amyotrophic lateral sclerosis (FTD/ALS)-associated RBPs TDP-43 and hnRNPA1 lead to distinct alterations in the neuritic proteome and transcriptome. TDP-43 knockdown (KD) leads to increased neuritic mRNA and translation. In contrast, hnRNPA1 leads to increased neuritic mRNA, but not translation, and more moderate effects on local mRNA profiles, possibly due to compensation by hnRNPA3. These results highlight the crucial role of FTD/ALS-associated RBPs in mRNA transport and local translation in neurons and the importance of these processes in neuron health and disease.
Collapse
Affiliation(s)
- Rachael Dargan
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Alla Mikheenko
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Nicholas L Johnson
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Benjamin Packer
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Ziyi Li
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Emma J Craig
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie L Sarbanes
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Colleen Bereda
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Puja R Mehta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Matthew Keuss
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Cory A Weller
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Pietro Fratta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
- Francis Crick Institute, London, UK
| | - Veronica H Ryan
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
32
|
van der Geest AT, Jakobs CE, Ljubikj T, Huffels CFM, Cañizares Luna M, Vieira de Sá R, Adolfs Y, de Wit M, Rutten DH, Kaal M, Zwartkruis MM, Carcolé M, Groen EJN, Hol EM, Basak O, Isaacs AM, Westeneng HJ, van den Berg LH, Veldink JH, Schlegel DK, Pasterkamp RJ. Molecular pathology, developmental changes and synaptic dysfunction in (pre-) symptomatic human C9ORF72-ALS/FTD cerebral organoids. Acta Neuropathol Commun 2024; 12:152. [PMID: 39289761 PMCID: PMC11409520 DOI: 10.1186/s40478-024-01857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
A hexanucleotide repeat expansion (HRE) in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Human brain imaging and experimental studies indicate early changes in brain structure and connectivity in C9-ALS/FTD, even before symptom onset. Because these early disease phenotypes remain incompletely understood, we generated iPSC-derived cerebral organoid models from C9-ALS/FTD patients, presymptomatic C9ORF72-HRE (C9-HRE) carriers, and controls. Our work revealed the presence of all three C9-HRE-related molecular pathologies and developmental stage-dependent size phenotypes in cerebral organoids from C9-ALS/FTD patients. In addition, single-cell RNA sequencing identified changes in cell type abundance and distribution in C9-ALS/FTD organoids, including a reduction in the number of deep layer cortical neurons and the distribution of neural progenitors. Further, molecular and cellular analyses and patch-clamp electrophysiology detected various changes in synapse structure and function. Intriguingly, organoids from all presymptomatic C9-HRE carriers displayed C9-HRE molecular pathology, whereas the extent to which more downstream cellular defects, as found in C9-ALS/FTD models, were detected varied for the different presymptomatic C9-HRE cases. Together, these results unveil early changes in 3D human brain tissue organization and synaptic connectivity in C9-ALS/FTD that likely constitute initial pathologies crucial for understanding disease onset and the design of therapeutic strategies.
Collapse
Affiliation(s)
- Astrid T van der Geest
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Channa E Jakobs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tijana Ljubikj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan F M Huffels
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Renata Vieira de Sá
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Daan H Rutten
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marthe Kaal
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maria M Zwartkruis
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Ewout J N Groen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henk-Jan Westeneng
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Domino K Schlegel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
33
|
Banack SA, Dunlop RA, Mehta P, Mitsumoto H, Wood SP, Han M, Cox PA. A microRNA diagnostic biomarker for amyotrophic lateral sclerosis. Brain Commun 2024; 6:fcae268. [PMID: 39280119 PMCID: PMC11398878 DOI: 10.1093/braincomms/fcae268] [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: 01/11/2024] [Revised: 06/13/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
Blood-based diagnostic biomarkers for amyotrophic lateral sclerosis will improve patient outcomes and positively impact novel drug development. Critical to the development of such biomarkers is robust method validation, optimization and replication with adequate sample sizes and neurological disease comparative blood samples. We sought to test an amyotrophic lateral sclerosis biomarker derived from diverse samples to determine if it is disease specific. Extracellular vesicles were extracted from blood plasma obtained from individuals diagnosed with amyotrophic lateral sclerosis, primary lateral sclerosis, Parkinson's disease and healthy controls. Immunoaffinity purification was used to create a neural-enriched extracellular vesicle fraction. MicroRNAs were measured across sample cohorts using real-time polymerase chain reaction. A Kruskal-Wallis test was used to assess differences in plasma microRNAs followed by post hoc Mann-Whitney tests to compare disease groups. Diagnostic accuracy was determined using a machine learning algorithm and a logistic regression model. We identified an eight-microRNA diagnostic signature for blood samples from amyotrophic lateral sclerosis patients with high sensitivity and specificity and an area under the curve calculation of 98% with clear statistical separation from neurological controls. The eight identified microRNAs represent disease-related biological processes consistent with amyotrophic lateral sclerosis. The direction and magnitude of gene fold regulation are consistent across four separate patient cohorts with real-time polymerase chain reaction analyses conducted in two laboratories from diverse samples and sample collection procedures. We propose that this diagnostic signature could be an aid to neurologists to supplement current clinical metrics used to diagnose amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
| | | | - Paul Mehta
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention, Atlanta, GA 30033, USA
| | - Hiroshi Mitsumoto
- Eleanor and Lou Gehrig MND/ALS Research Center, Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Moon Han
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention, Atlanta, GA 30033, USA
| | | |
Collapse
|
34
|
Huang WP, Ellis BCS, Hodgson RE, Sanchez Avila A, Kumar V, Rayment J, Moll T, Shelkovnikova TA. Stress-induced TDP-43 nuclear condensation causes splicing loss of function and STMN2 depletion. Cell Rep 2024; 43:114421. [PMID: 38941189 DOI: 10.1016/j.celrep.2024.114421] [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: 01/10/2024] [Revised: 04/04/2024] [Accepted: 06/14/2024] [Indexed: 06/30/2024] Open
Abstract
TDP-43 protein is dysregulated in several neurodegenerative diseases, which often have a multifactorial nature and may have extrinsic stressors as a "second hit." TDP-43 undergoes reversible nuclear condensation in stressed cells including neurons. Here, we demonstrate that stress-inducible nuclear TDP-43 condensates are RNA-depleted, non-liquid assemblies distinct from the known nuclear bodies. Their formation requires TDP-43 oligomerization and ATP and is inhibited by RNA. Using a confocal nanoscanning assay, we find that amyotrophic lateral sclerosis (ALS)-linked mutations alter stress-induced TDP-43 condensation by changing its affinity to liquid-like ribonucleoprotein assemblies. Stress-induced nuclear condensation transiently inactivates TDP-43, leading to loss of interaction with its protein binding partners and loss of function in splicing. Splicing changes are especially prominent and persisting for STMN2 RNA, and STMN2 protein becomes rapidly depleted early during stress. Our results point to early pathological changes to TDP-43 in the nucleus and support therapeutic modulation of stress response in ALS.
Collapse
Affiliation(s)
- Wan-Ping Huang
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Brittany C S Ellis
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Rachel E Hodgson
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Anna Sanchez Avila
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Vedanth Kumar
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Jessica Rayment
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Tobias Moll
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Tatyana A Shelkovnikova
- Sheffield Institute for Translational Neuroscience and Neuroscience Institute, University of Sheffield, Sheffield, UK.
| |
Collapse
|
35
|
Koike Y. Abnormal Splicing Events due to Loss of Nuclear Function of TDP-43: Pathophysiology and Perspectives. JMA J 2024; 7:313-318. [PMID: 39114608 PMCID: PMC11301021 DOI: 10.31662/jmaj.2024-0038] [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: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 08/10/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative diseases with a progressive and fatal course. They are often comorbid and share the same molecular spectrum. Their key pathological features are the formation of the aggregation of TDP-43, an RNA-binding protein, in the cytoplasm and its depletion from the nucleus in the central nervous system. In the nucleus, TDP-43 regulates several aspects of RNA metabolism, ranging from RNA transcription and alternative splicing to RNA transport. Suppressing the aberrant splicing events during RNA processing is one of the significant functions of TDP-43. This function is impaired when TDP-43 becomes depleted from the nucleus. Several critical cryptic splicing targets of TDP-43 have recently emerged, such as STMN2, UNC13A, and others. UNC13A is an important ALS/FTD risk gene, and the genetic variations, single nucleotide polymorphisms, cause disease via the increased susceptibility for cryptic exon inclusion under the TDP-43 dysfunction. Moreover, TDP-43 has an autoregulatory mechanism that regulates the splicing of its mRNA (TARDBP mRNA) in the healthy state. This study provides recent findings on the splicing regulatory function of TDP-43 and discusses the prospects of using these aberrant splicing events as efficient biomarkers.
Collapse
Affiliation(s)
- Yuka Koike
- Department of Molecular Neuroscience, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Neuroscience, Mayo Clinic Florida, Florida, USA
| |
Collapse
|
36
|
López-Carbonero JI, García-Toledo I, Fernández-Hernández L, Bascuñana P, Gil-Moreno MJ, Matías-Guiu JA, Corrochano S. In vivo diagnosis of TDP-43 proteinopathies: in search of biomarkers of clinical use. Transl Neurodegener 2024; 13:29. [PMID: 38831349 PMCID: PMC11149336 DOI: 10.1186/s40035-024-00419-8] [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: 01/16/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
TDP-43 proteinopathies are a heterogeneous group of neurodegenerative disorders that share the presence of aberrant, misfolded and mislocalized deposits of the protein TDP-43, as in the case of amyotrophic lateral sclerosis and some, but not all, pathological variants of frontotemporal dementia. In recent years, many other diseases have been reported to have primary or secondary TDP-43 proteinopathy, such as Alzheimer's disease, Huntington's disease or the recently described limbic-predominant age-related TDP-43 encephalopathy, highlighting the need for new and accurate methods for the early detection of TDP-43 proteinopathy to help on the stratification of patients with overlapping clinical diagnosis. Currently, TDP-43 proteinopathy remains a post-mortem pathologic diagnosis. Although the main aim is to determine the pathologic TDP-43 proteinopathy in the central nervous system (CNS), the ubiquitous expression of TDP-43 in biofluids and cells outside the CNS facilitates the use of other accessible target tissues that might reflect the potential TDP-43 alterations in the brain. In this review, we describe the main developments in the early detection of TDP-43 proteinopathies, and their potential implications on diagnosis and future treatments.
Collapse
Affiliation(s)
- Juan I López-Carbonero
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040, Madrid, Spain
| | - Irene García-Toledo
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040, Madrid, Spain
| | - Laura Fernández-Hernández
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040, Madrid, Spain
| | - Pablo Bascuñana
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040, Madrid, Spain
| | - María J Gil-Moreno
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040, Madrid, Spain
| | - Jordi A Matías-Guiu
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040, Madrid, Spain
| | - Silvia Corrochano
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040, Madrid, Spain.
| |
Collapse
|
37
|
Chatterjee M, Özdemir S, Fritz C, Möbius W, Kleineidam L, Mandelkow E, Biernat J, Doğdu C, Peters O, Cosma NC, Wang X, Schneider LS, Priller J, Spruth E, Kühn AA, Krause P, Klockgether T, Vogt IR, Kimmich O, Spottke A, Hoffmann DC, Fliessbach K, Miklitz C, McCormick C, Weydt P, Falkenburger B, Brandt M, Guenther R, Dinter E, Wiltfang J, Hansen N, Bähr M, Zerr I, Flöel A, Nestor PJ, Düzel E, Glanz W, Incesoy E, Bürger K, Janowitz D, Perneczky R, Rauchmann BS, Hopfner F, Wagemann O, Levin J, Teipel S, Kilimann I, Goerss D, Prudlo J, Gasser T, Brockmann K, Mengel D, Zimmermann M, Synofzik M, Wilke C, Selma-González J, Turon-Sans J, Santos-Santos MA, Alcolea D, Rubio-Guerra S, Fortea J, Carbayo Á, Lleó A, Rojas-García R, Illán-Gala I, Wagner M, Frommann I, Roeske S, Bertram L, Heneka MT, Brosseron F, Ramirez A, Schmid M, Beschorner R, Halle A, Herms J, Neumann M, Barthélemy NR, Bateman RJ, Rizzu P, Heutink P, Dols-Icardo O, Höglinger G, Hermann A, Schneider A. Plasma extracellular vesicle tau and TDP-43 as diagnostic biomarkers in FTD and ALS. Nat Med 2024; 30:1771-1783. [PMID: 38890531 PMCID: PMC11186765 DOI: 10.1038/s41591-024-02937-4] [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: 07/10/2023] [Accepted: 03/21/2024] [Indexed: 06/20/2024]
Abstract
Minimally invasive biomarkers are urgently needed to detect molecular pathology in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Here, we show that plasma extracellular vesicles (EVs) contain quantifiable amounts of TDP-43 and full-length tau, which allow the quantification of 3-repeat (3R) and 4-repeat (4R) tau isoforms. Plasma EV TDP-43 levels and EV 3R/4R tau ratios were determined in a cohort of 704 patients, including 37 genetically and 31 neuropathologically proven cases. Diagnostic groups comprised patients with TDP-43 proteinopathy ALS, 4R tauopathy progressive supranuclear palsy, behavior variant FTD (bvFTD) as a group with either tau or TDP-43 pathology, and healthy controls. EV tau ratios were low in progressive supranuclear palsy and high in bvFTD with tau pathology. EV TDP-43 levels were high in ALS and in bvFTD with TDP-43 pathology. Both markers discriminated between the diagnostic groups with area under the curve values >0.9, and between TDP-43 and tau pathology in bvFTD. Both markers strongly correlated with neurodegeneration, and clinical and neuropsychological markers of disease severity. Findings were replicated in an independent validation cohort of 292 patients including 34 genetically confirmed cases. Taken together, the combination of EV TDP-43 levels and EV 3R/4R tau ratios may aid the molecular diagnosis of FTD, FTD spectrum disorders and ALS, providing a potential biomarker to monitor disease progression and target engagement in clinical trials.
Collapse
Grants
- R01 AG080470 NIA NIH HHS
- This study was funded by a grant from the German Federal Ministry of Education and Research, BMBF, grant identifier 01KX2230 to AS. AS received funding from the Federal Ministry of Education and Research, BMBF (DESCARTES consortium, grant identifier 01EK2102A, and PREPARE, grant identifier 01GP2213A), Verum Foundation and BMBF/NUM (UTN consortium). A.S. received funding from Cure Alzheimer’s Fund and from Netzwerke NRW iBehave consortium. A.S. is member of the DFG-funded Cluster of Excellence ImmunoSensation2 - EXC2151 – 390873048. A.S. and A.R. were supported by La Fundación Reina Sofía, proyecto “MANOLO BARRÓS”. A.S. received funding by the Target ALS Foundation (TALS).
- MC received funding from Deutsche Demenzhilfe DZNE Innovative Minds Program and the Manfred-Strohscheer-Foundation.
- L.K. received funding from the Hertie Foundation, Hertie Network of Excellence in Clinical Neurosciences and from the JPND grant 01ED2007B (PreAdapt).
- Cure Alzheimer Foundation, Katharina Hard Foundation
- NRW Netzwerke iBehave
- DFG, Neuro-AcSis
Collapse
Affiliation(s)
| | - Selcuk Özdemir
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Genetics, Atatürk University, Erzurum, Turkey
| | - Christian Fritz
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Eckhard Mandelkow
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jacek Biernat
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Cem Doğdu
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | | | - Xiao Wang
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | | | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Technical University of Munich School of Medicine, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Eike Spruth
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea A Kühn
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Patricia Krause
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Klockgether
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Ina R Vogt
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Okka Kimmich
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | | | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Carolin Miklitz
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Cornelia McCormick
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Patrick Weydt
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Björn Falkenburger
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Moritz Brandt
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - René Guenther
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Elisabeth Dinter
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Niels Hansen
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Mathias Bähr
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
- Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Göttingen, Germany
| | - Inga Zerr
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
| | - Peter J Nestor
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Queensland Brain Institute, University of Queensland and Mater Public Hospital, Brisbane, Queensland, Australia
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Clinic for Neurology, University Hospital Magdeburg, Magdeburg, Germany
| | - Enise Incesoy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Magdeburg, Magdeburg, Germany
| | - Katharina Bürger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Boris S Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Department of Neuroradiology, University Hospital LMU, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Franziska Hopfner
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Olivia Wagemann
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Stefan Teipel
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Ingo Kilimann
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Doreen Goerss
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
| | - Johannes Prudlo
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
- Department of Neurology, Rostock University Medical Centre, Rostock, Germany
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Kathrin Brockmann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - David Mengel
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Milan Zimmermann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Carlo Wilke
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Judit Selma-González
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Motor Neuron Disease Clinic, Neuromuscular Diseases Unit, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Janina Turon-Sans
- Motor Neuron Disease Clinic, Neuromuscular Diseases Unit, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Miguel Angel Santos-Santos
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Sara Rubio-Guerra
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Álvaro Carbayo
- Motor Neuron Disease Clinic, Neuromuscular Diseases Unit, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ricardo Rojas-García
- Motor Neuron Disease Clinic, Neuromuscular Diseases Unit, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Ingo Frommann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Sandra Roeske
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lucas Bertram
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachussetss Medical School, North Worcester, MA, USA
| | | | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry, University of Cologne, Cologne, Germany
- Department of Psychiatry, Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Matthias Schmid
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Rudi Beschorner
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, University of Tübingen, Tübingen, Germany
| | - Annett Halle
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Manuela Neumann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, University of Tübingen, Tübingen, Germany
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Andreas Hermann
- German Centre for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
- Translational Neurodegeneration Section 'Albrecht Kossel' and Center for Transdisciplinary Neurosciences, University Medical Center Rostock, Rostock, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
38
|
Benatar M, Wuu J, Huey ED, McMillan CT, Petersen RC, Postuma R, McHutchison C, Dratch L, Arias JJ, Crawley A, Houlden H, McDermott MP, Cai X, Thakur N, Boxer A, Rosen H, Boeve BF, Dacks P, Cosentino S, Abrahams S, Shneider N, Lingor P, Shefner J, Andersen PM, Al-Chalabi A, Turner MR. The Miami Framework for ALS and related neurodegenerative disorders: an integrated view of phenotype and biology. Nat Rev Neurol 2024; 20:364-376. [PMID: 38769202 PMCID: PMC11216694 DOI: 10.1038/s41582-024-00961-z] [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] [Accepted: 04/03/2024] [Indexed: 05/22/2024]
Abstract
Increasing appreciation of the phenotypic and biological overlap between amyotrophic lateral sclerosis (ALS) and frontotemporal dementia, alongside evolving biomarker evidence for a pre-symptomatic stage of disease and observations that this stage of disease might not always be clinically silent, is challenging traditional views of these disorders. These advances have highlighted the need to adapt ingrained notions of these clinical syndromes to include both the full phenotypic continuum - from clinically silent, to prodromal, to clinically manifest - and the expanded phenotypic spectrum that includes ALS, frontotemporal dementia and some movement disorders. The updated clinical paradigms should also align with our understanding of the biology of these disorders, reflected in measurable biomarkers. The Miami Framework, emerging from discussions at the Second International Pre-Symptomatic ALS Workshop in Miami (February 2023; a full list of attendees and their affiliations appears in the Supplementary Information) proposes a classification system built on: first, three parallel phenotypic axes - motor neuron, frontotemporal and extrapyramidal - rather than the unitary approach of combining all phenotypic elements into a single clinical entity; and second, biomarkers that reflect different aspects of the underlying pathology and biology of neurodegeneration. This framework decouples clinical syndromes from biomarker evidence of disease and builds on experiences from other neurodegenerative diseases to offer a unified approach to specifying the pleiotropic clinical manifestations of disease and describing the trajectory of emergent biomarkers.
Collapse
Affiliation(s)
- Michael Benatar
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Joanne Wuu
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Edward D Huey
- Department of Psychiatry and Human Behaviour, Alpert Medical School of Brown University, Providence, RI, USA
| | - Corey T McMillan
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Ronald Postuma
- Department of Neurology, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Caroline McHutchison
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Laynie Dratch
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jalayne J Arias
- Department of Health Policy & Behavioral Sciences, School of Public Health, Georgia State University, Atlanta, GA, USA
| | | | - Henry Houlden
- UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Michael P McDermott
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xueya Cai
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Adam Boxer
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Howard Rosen
- Department of Neurology, University of California, San Francisco, CA, USA
| | | | - Penny Dacks
- Association for Frontotemporal Degeneration, King of Prussia, PA, USA
| | | | - Sharon Abrahams
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Neil Shneider
- Department of Neurology, Columbia University, New York, NY, USA
| | - Paul Lingor
- Department of Neurology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Jeremy Shefner
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Peter M Andersen
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- Department of Neurology, King's College Hospital, London, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
39
|
Van Es MA. Amyotrophic lateral sclerosis; clinical features, differential diagnosis and pathology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:1-47. [PMID: 38802173 DOI: 10.1016/bs.irn.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset syndrome characterized by the progressive degeneration of both upper motor neurons (UMN) and lower motor neurons (LMN). ALS forms a clinical continuum with frontotemporal dementia (FTD), in which there are progressive language deficits or behavioral changes. The genetics and pathology underlying both ALS and FTD overlap as well, with cytoplasmatic misvocalization of TDP-43 as the hallmark. ALS is diagnosed by exclusion. Over the years several diagnostic criteria have been proposed, which in essence all require a history of slowly progressive motor symptoms, with UMN and LMN signs on neurological examination, clear spread of symptoms through the body, the exclusion of other disorder that cause similar symptoms and an EMG that it is compatible with LMN loss. ALS is heterogeneous disorder that may present in multitude ways, which makes the diagnosis challenging. Therefore, a systematic approach in the diagnostic process is required in line with the most common presentations. Subsequently, assessing whether there are cognitive and/or behavioral changes within the spectrum of FTD and lastly determining the cause is genetic. This chapter, an outline on how to navigate this 3 step process.
Collapse
Affiliation(s)
- Michael A Van Es
- Department of Neurology, Brain Center UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
40
|
Nelson PT, Fardo DW, Wu X, Aung KZ, Cykowski MD, Katsumata Y. Limbic-predominant age-related TDP-43 encephalopathy (LATE-NC): Co-pathologies and genetic risk factors provide clues about pathogenesis. J Neuropathol Exp Neurol 2024; 83:396-415. [PMID: 38613823 PMCID: PMC11110076 DOI: 10.1093/jnen/nlae032] [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] [Indexed: 04/15/2024] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is detectable at autopsy in more than one-third of people beyond age 85 years and is robustly associated with dementia independent of other pathologies. Although LATE-NC has a large impact on public health, there remain uncertainties about the underlying biologic mechanisms. Here, we review the literature from human studies that may shed light on pathogenetic mechanisms. It is increasingly clear that certain combinations of pathologic changes tend to coexist in aging brains. Although "pure" LATE-NC is not rare, LATE-NC often coexists in the same brains with Alzheimer disease neuropathologic change, brain arteriolosclerosis, hippocampal sclerosis of aging, and/or age-related tau astrogliopathy (ARTAG). The patterns of pathologic comorbidities provide circumstantial evidence of mechanistic interactions ("synergies") between the pathologies, and also suggest common upstream influences. As to primary mediators of vulnerability to neuropathologic changes, genetics may play key roles. Genes associated with LATE-NC include TMEM106B, GRN, APOE, SORL1, ABCC9, and others. Although the anatomic distribution of TDP-43 pathology defines the condition, important cofactors for LATE-NC may include Tau pathology, endolysosomal pathways, and blood-brain barrier dysfunction. A review of the human phenomenology offers insights into disease-driving mechanisms, and may provide clues for diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - David W Fardo
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Xian Wu
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Khine Zin Aung
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Yuriko Katsumata
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
41
|
Choi HK, Chen M, Goldston LL, Lee KB. Extracellular vesicles as nanotheranostic platforms for targeted neurological disorder interventions. NANO CONVERGENCE 2024; 11:19. [PMID: 38739358 PMCID: PMC11091041 DOI: 10.1186/s40580-024-00426-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/24/2024] [Indexed: 05/14/2024]
Abstract
Central Nervous System (CNS) disorders represent a profound public health challenge that affects millions of people around the world. Diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and traumatic brain injury (TBI) exemplify the complexities and diversities that complicate their early detection and the development of effective treatments. Amid these challenges, the emergence of nanotechnology and extracellular vesicles (EVs) signals a new dawn for treating and diagnosing CNS ailments. EVs are cellularly derived lipid bilayer nanosized particles that are pivotal in intercellular communication within the CNS and have the potential to revolutionize targeted therapeutic delivery and the identification of novel biomarkers. Integrating EVs with nanotechnology amplifies their diagnostic and therapeutic capabilities, opening new avenues for managing CNS diseases. This review focuses on examining the fascinating interplay between EVs and nanotechnology in CNS theranostics. Through highlighting the remarkable advancements and unique methodologies, we aim to offer valuable perspectives on how these approaches can bring about a revolutionary change in disease management. The objective is to harness the distinctive attributes of EVs and nanotechnology to forge personalized, efficient interventions for CNS disorders, thereby providing a beacon of hope for affected individuals. In short, the confluence of EVs and nanotechnology heralds a promising frontier for targeted and impactful treatments against CNS diseases, which continue to pose significant public health challenges. By focusing on personalized and powerful diagnostic and therapeutic methods, we might improve the quality of patients.
Collapse
Affiliation(s)
- Hye Kyu Choi
- Department of Chemistry and Chemical Biology, The State University of New Jersey, 123 Bevier Road, Rutgers, Piscataway, NJ, 08854, USA
| | - Meizi Chen
- Department of Chemistry and Chemical Biology, The State University of New Jersey, 123 Bevier Road, Rutgers, Piscataway, NJ, 08854, USA
| | - Li Ling Goldston
- Department of Chemistry and Chemical Biology, The State University of New Jersey, 123 Bevier Road, Rutgers, Piscataway, NJ, 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, The State University of New Jersey, 123 Bevier Road, Rutgers, Piscataway, NJ, 08854, USA.
| |
Collapse
|
42
|
Eisen A, Pioro EP, Goutman SA, Kiernan MC. Nanoplastics and Neurodegeneration in ALS. Brain Sci 2024; 14:471. [PMID: 38790450 PMCID: PMC11119293 DOI: 10.3390/brainsci14050471] [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: 04/18/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Plastic production, which exceeds one million tons per year, is of global concern. The constituent low-density polymers enable spread over large distances and micro/nano particles (MNPLs) induce organ toxicity via digestion, inhalation, and skin contact. Particles have been documented in all human tissues including breast milk. MNPLs, especially weathered particles, can breach the blood-brain barrier, inducing neurotoxicity. This has been documented in non-human species, and in human-induced pluripotent stem cell lines. Within the brain, MNPLs initiate an inflammatory response with pro-inflammatory cytokine production, oxidative stress with generation of reactive oxygen species, and mitochondrial dysfunction. Glutamate and GABA neurotransmitter dysfunction also ensues with alteration of excitatory/inhibitory balance in favor of reduced inhibition and resultant neuro-excitation. Inflammation and cortical hyperexcitability are key abnormalities involved in the pathogenic cascade of amyotrophic lateral sclerosis (ALS) and are intricately related to the mislocalization and aggregation of TDP-43, a hallmark of ALS. Water and many foods contain MNPLs and in humans, ingestion is the main form of exposure. Digestion of plastics within the gut can alter their properties, rendering them more toxic, and they cause gut microbiome dysbiosis and a dysfunctional gut-brain axis. This is recognized as a trigger and/or aggravating factor for ALS. ALS is associated with a long (years or decades) preclinical period and neonates and infants are exposed to MNPLs through breast milk, milk substitutes, and toys. This endangers a time of intense neurogenesis and establishment of neuronal circuitry, setting the stage for development of neurodegeneration in later life. MNPL neurotoxicity should be considered as a yet unrecognized risk factor for ALS and related diseases.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6S 1Z3, Canada;
| | - Erik P. Pioro
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6S 1Z3, Canada;
| | - Stephen A. Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA;
| | | |
Collapse
|
43
|
Calliari A, Daughrity LM, Albagli EA, Castellanos Otero P, Yue M, Jansen-West K, Islam NN, Caulfield T, Rawlinson B, DeTure M, Cook C, Graff-Radford NR, Day GS, Boeve BF, Knopman DS, Petersen RC, Josephs KA, Oskarsson B, Gitler AD, Dickson DW, Gendron TF, Prudencio M, Ward ME, Zhang YJ, Petrucelli L. HDGFL2 cryptic proteins report presence of TDP-43 pathology in neurodegenerative diseases. Mol Neurodegener 2024; 19:29. [PMID: 38539264 PMCID: PMC10967196 DOI: 10.1186/s13024-024-00718-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
This letter demonstrates the potential of novel cryptic proteins resulting from TAR DNA-binding protein 43 (TDP-43) dysfunction as markers of TDP-43 pathology in neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Calliari
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ellen A Albagli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Mei Yue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Naeyma N Islam
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Casey Cook
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | | | | | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mercedes Prudencio
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
44
|
Cheslow L, Snook AE, Waldman SA. Biomarkers for Managing Neurodegenerative Diseases. Biomolecules 2024; 14:398. [PMID: 38672416 PMCID: PMC11048498 DOI: 10.3390/biom14040398] [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: 03/03/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Neurological disorders are the leading cause of cognitive and physical disability worldwide, affecting 15% of the global population. Due to the demographics of aging, the prevalence of neurological disorders, including neurodegenerative diseases, will double over the next two decades. Unfortunately, while available therapies provide symptomatic relief for cognitive and motor impairment, there is an urgent unmet need to develop disease-modifying therapies that slow the rate of pathological progression. In that context, biomarkers could identify at-risk and prodromal patients, monitor disease progression, track responses to therapy, and parse the causality of molecular events to identify novel targets for further clinical investigation. Thus, identifying biomarkers that discriminate between diseases and reflect specific stages of pathology would catalyze the discovery and development of therapeutic targets. This review will describe the prevalence, known mechanisms, ongoing or recently concluded therapeutic clinical trials, and biomarkers of three of the most prevalent neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD).
Collapse
Affiliation(s)
- Lara Cheslow
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.C.); (A.E.S.)
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E. Snook
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.C.); (A.E.S.)
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Scott A. Waldman
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.C.); (A.E.S.)
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Frontotemporal dementia (FTD) is a clinically, pathologically and genetically heterogeneous disorder. Whilst disease modifying therapy trials are mostly focused on the symptomatic phase, future studies will move earlier in the disease aiming to prevent symptom onset. This review summarizes the recent work to better understand this presymptomatic period. RECENT FINDINGS The presymptomatic phase can be split into preclinical and prodromal stages. The onset of the preclinical phase is defined by the first presence of pathological inclusions of tau, TDP-43 or fused in sarcoma in the brain. Definitive biomarkers of these pathologies do not yet exist for FTD. The prodromal phase is defined by the onset of mild symptoms. Recent work has highlighted the wide phenotypic spectrum that occurs, with the concept of mild cognitive ± behavioural ± motor impairment (MCBMI) being put forward, and additions to scales such as the CDR plus NACC FTLD now incorporating neuropsychiatric and motor symptoms. SUMMARY It will be important to better characterize the presymptomatic period moving forward and develop robust biomarkers that can be used both for stratification and outcome measures in prevention trials. The work of the FTD Prevention Initiative aims to facilitate this by bringing together data from natural history studies across the world.
Collapse
Affiliation(s)
- Lucy L Russell
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | |
Collapse
|