1
|
Uliassi E, Bolognesi ML, Milelli A. Targeting Tau Protein with Proximity Inducing Modulators: A New Frontier to Combat Tauopathies. ACS Pharmacol Transl Sci 2025; 8:654-672. [PMID: 40109749 PMCID: PMC11915046 DOI: 10.1021/acsptsci.4c00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 03/22/2025]
Abstract
Dysregulation of correct protein tau homeostasis represents the seed for the development of several devastating central nervous system disorders, known as tauopathies, that affect millions of people worldwide. Despite massive public and private support to research funding, these diseases still represent unmet medical needs. In fact, the tau-targeting tools developed to date have failed to translate into the clinic. Recently, taking advantage of the modes that nature uses to mediate the flow of information in cells, researchers have developed a new class of molecules, called proximity-inducing modulators, which exploit spatial proximity to modulate protein function(s) and redirect cellular processes. In this perspective, after a brief discussion about tau protein and the classic tau-targeting approaches, we will discuss the different classes of proximity-inducing modulators developed so far and highlight the applications to modulate tau protein's function and tau-induced toxicity.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Andrea Milelli
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d'Augusto 237, Rimini 47921, Italy
| |
Collapse
|
2
|
Cummings JL, Zhou Y, Van Stone A, Cammann D, Tonegawa-Kuji R, Fonseca J, Cheng F. Drug repurposing for Alzheimer's disease and other neurodegenerative disorders. Nat Commun 2025; 16:1755. [PMID: 39971900 PMCID: PMC11840136 DOI: 10.1038/s41467-025-56690-4] [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: 04/17/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Repurposed drugs provide a rich source of potential therapies for Alzheimer's disease (AD) and other neurodegenerative disorders (NDD). Repurposed drugs have information from non-clinical studies, phase 1 dosing, and safety and tolerability data collected with the original indication. Computational approaches, "omic" studies, drug databases, and electronic medical records help identify candidate therapies. Generic repurposed agents lack intellectual property protection and are rarely advanced to late-stage trials for AD/NDD. In this review we define repurposing, describe the advantages and challenges of repurposing, offer strategies for overcoming the obstacles, and describe the key contributions of repurposing to the drug development ecosystem.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89106, USA.
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | | | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Reina Tonegawa-Kuji
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | - Jorge Fonseca
- Howard R Hughes College of Engineering, Department of Computer Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89154, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106, USA
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| |
Collapse
|
3
|
Scotton WJ, Shand C, Todd EG, Bocchetta M, Kobylecki C, Cash DM, VandeVrede L, Heuer HW, Quaegebeur A, Young AL, Oxtoby N, Alexander D, Rowe JB, Morris HR, Boxer AL, Rohrer JD, Wijeratne PA. Distinct spatiotemporal atrophy patterns in corticobasal syndrome are associated with different underlying pathologies. Brain Commun 2025; 7:fcaf066. [PMID: 40070441 PMCID: PMC11894806 DOI: 10.1093/braincomms/fcaf066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 01/02/2025] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Although the corticobasal syndrome was originally most closely linked with the pathology of corticobasal degeneration, the 2013 Armstrong clinical diagnostic criteria, without the addition of aetiology-specific biomarkers, have limited positive predictive value for identifying corticobasal degeneration pathology in life. Autopsy studies demonstrate considerable pathological heterogeneity in corticobasal syndrome, with corticobasal degeneration pathology accounting for only ∼50% of clinically diagnosed individuals. Individualized disease stage and progression modelling of brain changes in corticobasal syndrome may have utility in predicting this underlying pathological heterogeneity, and in turn improve the design of clinical trials for emerging disease-modifying therapies. The aim of this study was to jointly model the phenotypic and temporal heterogeneity of corticobasal syndrome, to identify unique imaging subtypes based solely on a data-driven assessment of MRI atrophy patterns and then investigate whether these subtypes provide information on the underlying pathology. We applied Subtype and Stage Inference, a machine learning algorithm that identifies groups of individuals with distinct biomarker progression patterns, to a large cohort of 135 individuals with corticobasal syndrome (52 had a pathological or biomarker defined diagnosis) and 252 controls. The model was fit using volumetric features extracted from baseline T1-weighted MRI scans and then used to subtype and stage follow-up scans. The subtypes and stages at follow-up were used to validate the longitudinal consistency of the baseline subtype and stage assignments. We then investigated whether there were differences in associated pathology and clinical phenotype between the subtypes. Subtype and Stage Inference identified at least two distinct and longitudinally stable spatiotemporal subtypes of atrophy progression in corticobasal syndrome; four-repeat-tauopathy confirmed cases were most commonly assigned to the Subcortical subtype (83% of individuals with progressive supranuclear palsy pathology and 75% of individuals with corticobasal-degeneration pathology), whilst those with Alzheimer's pathology were most commonly assigned to the Fronto-parieto-occipital subtype (81% of individuals). Subtype assignment was stable at follow-up (98% of cases), and individuals consistently progressed to higher stages (100% stayed at the same stage or progressed), supporting the model's ability to stage progression. By jointly modelling disease stage and subtype, we provide data-driven evidence for at least two distinct and longitudinally stable spatiotemporal subtypes of atrophy in corticobasal syndrome that are associated with different underlying pathologies. In the absence of sensitive and specific biomarkers, accurately subtyping and staging individuals with corticobasal syndrome at baseline has important implications for screening on entry into clinical trials, as well as for tracking disease progression.
Collapse
Affiliation(s)
- William J Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Cameron Shand
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
- Centre for Cognitive and Clinical Neuroscience, Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University, London UB8 3PH, UK
| | - Christopher Kobylecki
- Department of Neurology, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust (Salford Royal Hospital), Salford M6 8HD, UK
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Lawren VandeVrede
- Department of Neurology, Memory and Aging Center, University of California, San Francisco CA 94158, USA
| | - Hilary W Heuer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco CA 94158, USA
| | - Annelies Quaegebeur
- Cambridge University Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, Cambridge CB2 0QQ, UK
- Cambridge Brain Bank, Cambridge CB2 0QQ, UK
| | - Alexandra L Young
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| | - Neil Oxtoby
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| | - Daniel Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| | - James B Rowe
- Cambridge University Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, Cambridge CB2 0QQ, UK
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- Movement Disorders Centre, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco CA 94158, USA
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Peter A Wijeratne
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
- Department of Informatics, University of Sussex, Brighton BN1 9RH, UK
| |
Collapse
|
4
|
Singh S, Khan S, Shahid M, Sardar M, Hassan MI, Islam A. Targeting tau in Alzheimer's and beyond: Insights into pathology and therapeutic strategies. Ageing Res Rev 2025; 104:102639. [PMID: 39674375 DOI: 10.1016/j.arr.2024.102639] [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: 10/14/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Tauopathies encompass a group of approximately 20 neurodegenerative diseases characterized by the accumulation of the microtubule-associated protein tau in brain neurons. The pathogenesis of intracellular neurofibrillary tangles, a hallmark of tauopathies, is initiated by hyperphosphorylated tau protein isoforms that cause neuronal death and lead to diseases like Alzheimer's, Parkinson's disease, frontotemporal dementia, and other complex neurodegenerative diseases. Current applications of tau biomarkers, including imaging, cerebrospinal fluid, and blood-based assays, assist in the evaluation and diagnosis of tauopathies. Emerging research is providing various potential strategies to prevent cellular toxicity caused by tau aggregation such as: 1) suppressing toxic tau aggregation, 2) preventing post-translational modifications of tau, 3) stabilizing microtubules and 4) designing tau-directed immunogens. This review aims to discuss the role of tau in tauopathies along with neuropathological features of the different tauopathies and the new developments in treating tau aggregation with the therapeutics for treating and possibly preventing tauopathies.
Collapse
Affiliation(s)
- Sunidhi Singh
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Sumaiya Khan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Meryam Sardar
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
5
|
Marotta C, Sinclair B, O’Brien TJ, Vivash L. Biomarkers of disease progression in progressive supranuclear palsy for use in clinical trials. Brain Commun 2025; 7:fcaf022. [PMID: 39882025 PMCID: PMC11775610 DOI: 10.1093/braincomms/fcaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/10/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025] Open
Abstract
Progressive supranuclear palsy (PSP) is a rare neurodegenerative disease with no current disease-modifying treatments approved. Longitudinal research and clinical trials for PSP are ongoing and require reliable measures that are sensitive to disease progression. Despite susceptibility to subjective limitations, clinical and cognitive assessments are the most used instruments in therapeutic trials in PSP. The objective of this review was to identify measures that have been studied longitudinally as measures of progression and are suitable for use as clinical trial endpoints. We reviewed the measures currently used as trial endpoints, identifying the clinical, cognitive, fluid and imaging measures that have previously been studied longitudinally, and discuss current diagnostic and emerging measures that are yet to be studied longitudinally but that may be sensitive to disease progression. We found that many fluid and imaging measures require further research to validate their use as longitudinal measures of change, including emerging measures that have not yet been studied specifically in PSP. We also summarize the sample size estimates required to detect changes in a two-arm, 52-week therapeutic trial and found that specific MRI volumes require the smallest sample sizes to detect change.
Collapse
Affiliation(s)
- Cassandra Marotta
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Benjamin Sinclair
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Terence J O’Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Lucy Vivash
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| |
Collapse
|
6
|
Rajbanshi B, Prufer Q C Araujo I, VandeVrede L, Ljubenkov PA, Staffaroni AM, Heuer HW, Lario Lago A, Ramos EM, Petrucelli L, Gendron T, Dage JL, Seeley WW, Grinberg LT, Spina S, Bateman RJ, Rosen HJ, Boeve BF, Boxer AL, Rojas JC. Clinical and neuropathological associations of plasma Aβ 42/Aβ 40, p-tau217 and neurofilament light in sporadic frontotemporal dementia spectrum disorders. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70078. [PMID: 39886325 PMCID: PMC11780117 DOI: 10.1002/dad2.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 02/01/2025]
Abstract
INTRODUCTION Plasma amyloid beta42/amyloid beta40 (Aβ42/Aβ40) and phosphorylated tau217 (p-tau217) identify individuals with primary Alzheimer's disease (AD). They may detect AD co-pathology in the setting of other primary neurodegenerative diseases, but this has not been systematically studied. METHODS We compared the clinical, neuroimaging, and neuropathological associations of plasma Aβ42/Aβ40 (mass spectrometry), p-tau217 (electrochemiluminescence), and neurofilament light ([NfL], single molecule array [Simoa]), as markers of AD co-pathology, in a sporadic frontotemporal dementia (FTD) cohort (n = 620). RESULTS Aβ42/Aβ40 showed no clinicopathological associations. High p-tau217 was present in amnestic dementia (AmD) presumed to be due to FTD, logopenic primary progressive aphasia (lvPPA), and APOEε4 carriers, and correlated with worse baseline and longitudinal clinical scores, lower hippocampal volumes, and more severe AD co-pathology (Braak Stage). NfL was elevated in all FTD phenotypes, and correlated with clinical scores and frontotemporal brain volumes. DISCUSSION Plasma p-tau217 has clinical, neuroimaging, and neuropathological correlates in sporadic FTD and may identify FTD cases with AD co-pathology. Highlights Alzheimer's disease (AD) features could be identified with plasma phosphorylated tau217 (p-tau217) in frontotemporal lobar degeneration (FTLD).Plasma p-tau217 is a better discriminator of AD co-pathology and AD-associated features in FTLD than plasma amyloid beta42/amyloid beta40 (Aβ42/Aβ40) and neurofilament light (NfL).In FTLD, plasma p-tau217, but not Aβ42/Aβ40 or neurofilament light, has phenotypical, neurocognitive, and neuroimaging correlates suggestive of AD co-pathology.
Collapse
Affiliation(s)
- Binita Rajbanshi
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Igor Prufer Q C Araujo
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lawren VandeVrede
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Peter A. Ljubenkov
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Adam M. Staffaroni
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Hilary W. Heuer
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Argentina Lario Lago
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Eliana Marisa Ramos
- Department of Neurology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | | | - Tania Gendron
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Jeffrey L. Dage
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - William W. Seeley
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lea T. Grinberg
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Salvatore Spina
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Howard J. Rosen
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Adam L. Boxer
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Julio C. Rojas
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | |
Collapse
|
7
|
Zhang X, Wang J, Zhang Z, Ye K. Tau in neurodegenerative diseases: molecular mechanisms, biomarkers, and therapeutic strategies. Transl Neurodegener 2024; 13:40. [PMID: 39107835 PMCID: PMC11302116 DOI: 10.1186/s40035-024-00429-6] [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: 11/22/2023] [Accepted: 07/05/2024] [Indexed: 09/14/2024] Open
Abstract
The deposition of abnormal tau protein is characteristic of Alzheimer's disease (AD) and a class of neurodegenerative diseases called tauopathies. Physiologically, tau maintains an intrinsically disordered structure and plays diverse roles in neurons. Pathologically, tau undergoes abnormal post-translational modifications and forms oligomers or fibrous aggregates in tauopathies. In this review, we briefly introduce several tauopathies and discuss the mechanisms mediating tau aggregation and propagation. We also describe the toxicity of tau pathology. Finally, we explore the early diagnostic biomarkers and treatments targeting tau. Although some encouraging results have been achieved in animal experiments and preclinical studies, there is still no cure for tauopathies. More in-depth basic and clinical research on the pathogenesis of tauopathies is necessary.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiangyu Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
8
|
Dunning EE, Decourt B, Zawia NH, Shill HA, Sabbagh MN. Pharmacotherapies for the Treatment of Progressive Supranuclear Palsy: A Narrative Review. Neurol Ther 2024; 13:975-1013. [PMID: 38743312 PMCID: PMC11263316 DOI: 10.1007/s40120-024-00614-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disorder resulting from the deposition of misfolded and neurotoxic forms of tau protein in specific areas of the midbrain, basal ganglia, and cortex. It is one of the most representative forms of tauopathy. PSP presents in several different phenotypic variations and is often accompanied by the development of concurrent neurodegenerative disorders. PSP is universally fatal, and effective disease-modifying therapies for PSP have not yet been identified. Several tau-targeting treatment modalities, including vaccines, monoclonal antibodies, and microtubule-stabilizing agents, have been investigated and have had no efficacy. The need to treat PSP and other tauopathies is critical, and many clinical trials investigating tau-targeted treatments are underway. In this review, the PubMed database was queried to collect information about preclinical and clinical research on PSP treatment. Additionally, the US National Library of Medicine's ClinicalTrials.gov website was queried to identify past and ongoing clinical trials relevant to PSP treatment. This narrative review summarizes our findings regarding these reports, which include potential disease-modifying drug trials, modifiable risk factor management, and symptom treatments.
Collapse
Affiliation(s)
- Elise E Dunning
- Creighton University School of Medicine - Phoenix, Phoenix, AZ, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Laboratory on Neurodegeneration and Translational Research, College of Medicine, Roseman University of Health Sciences, Las Vegas, NV, USA
| | - Nasser H Zawia
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
- Department of Biomedical and Pharmaceutical Sciences, Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
| | - Holly A Shill
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | - Marwan N Sabbagh
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
9
|
Cardim-Pires TR, de Rus Jacquet A, Cicchetti F. Healthy blood, healthy brain: a window into understanding and treating neurodegenerative diseases. J Neurol 2024; 271:3682-3689. [PMID: 38607433 DOI: 10.1007/s00415-024-12337-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024]
Abstract
Our limited understanding of complex neurodegenerative disorders has held us back on the development of efficient therapies. While several approaches are currently being considered, it is still unclear what will be most successful. Among the latest and more novel ideas, the concept of blood or plasma transfusion from young healthy donors to diseased patients is gaining momentum and attracting attention beyond the scientific arena. While young or healthy blood is enriched with protective and restorative components, blood from older subjects may accumulate neurotoxic agents or be impoverished of beneficial factors. In this commentary, we present an overview of the compelling evidence collected in various animal models of brain diseases (e.g., Alzheimer, Parkinson, Huntington) to the actual clinical trials that have been conducted to test the validity of blood-related treatments in neurodegenerative diseases and argue in favor of such approach.
Collapse
Affiliation(s)
- Thyago R Cardim-Pires
- Centre de Recherche du CHU de Québec, Université Laval, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC, G1V 4G2, Canada
| | - Aurélie de Rus Jacquet
- Centre de Recherche du CHU de Québec, Université Laval, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC, G1V 4G2, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1K 0A6, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Université Laval, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC, G1V 4G2, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1K 0A6, Canada.
| |
Collapse
|
10
|
Singh H, Das A, Khan MM, Pourmotabbed T. New insights into the therapeutic approaches for the treatment of tauopathies. Neural Regen Res 2024; 19:1020-1026. [PMID: 37862204 PMCID: PMC10749630 DOI: 10.4103/1673-5374.385288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/05/2023] [Accepted: 08/10/2023] [Indexed: 10/22/2023] Open
Abstract
Tauopathies are a group of neurological disorders, including Alzheimer's disease and frontotemporal dementia, which involve progressive neurodegeneration, cognitive deficits, and aberrant tau protein accumulation. The development of tauopathies cannot currently be stopped or slowed down by treatment measures. Given the significant contribution of tau burden in primary tauopathies and the strong association between pathogenic tau accumulation and cognitive deficits, there has been a lot of interest in creating therapies that can alleviate tau pathology and render neuroprotective effects. Recently, small molecules, immunotherapies, and gene therapy have been used to reduce the pathological tau burden and prevent neurodegeneration in animal models of tauopathies. However, the major pitfall of the current therapeutic approach is the difficulty of drugs and gene-targeting modalities to cross the blood-brain barrier and their unintended side effects. In this review, the current therapeutic strategies used for tauopathies including the use of oligonucleotide-based gene therapy approaches that have shown a promising result for the treatment of tauopathies and Alzheimer's disease in preclinical animal models, have been discussed.
Collapse
Affiliation(s)
- Himanshi Singh
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India
| | - Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tayebeh Pourmotabbed
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
11
|
Buchholz S, Zempel H. The six brain-specific TAU isoforms and their role in Alzheimer's disease and related neurodegenerative dementia syndromes. Alzheimers Dement 2024; 20:3606-3628. [PMID: 38556838 PMCID: PMC11095451 DOI: 10.1002/alz.13784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 04/02/2024]
Abstract
INTRODUCTION Alternative splicing of the human MAPT gene generates six brain-specific TAU isoforms. Imbalances in the TAU isoform ratio can lead to neurodegenerative diseases, underscoring the need for precise control over TAU isoform balance. Tauopathies, characterized by intracellular aggregates of hyperphosphorylated TAU, exhibit extensive neurodegeneration and can be classified by the TAU isoforms present in pathological accumulations. METHODS A comprehensive review of TAU and related dementia syndromes literature was conducted using PubMed, Google Scholar, and preprint server. RESULTS While TAU is recognized as key driver of neurodegeneration in specific tauopathies, the contribution of the isoforms to neuronal function and disease development remains largely elusive. DISCUSSION In this review we describe the role of TAU isoforms in health and disease, and stress the importance of comprehending and studying TAU isoforms in both, physiological and pathological context, in order to develop targeted therapeutic interventions for TAU-associated diseases. HIGHLIGHTS MAPT splicing is tightly regulated during neuronal maturation and throughout life. TAU isoform expression is development-, cell-type and brain region specific. The contribution of TAU to neurodegeneration might be isoform-specific. Ineffective TAU-based therapies highlight the need for specific targeting strategies.
Collapse
Affiliation(s)
- Sarah Buchholz
- Institute of Human GeneticsFaculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Present address:
Department Schaefer, Neurobiology of AgeingMax Planck Institute for Biology of AgeingCologneGermany
| | - Hans Zempel
- Institute of Human GeneticsFaculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| |
Collapse
|
12
|
Scotton WJ, Shand C, Todd EG, Bocchetta M, Cash DM, VandeVrede L, Heuer HW, Young AL, Oxtoby N, Alexander DC, Rowe JB, Morris HR, Boxer AL, Rohrer JD, Wijeratne PA. Distinct spatiotemporal atrophy patterns in corticobasal syndrome are associated with different underlying pathologies. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.14.24304298. [PMID: 38562801 PMCID: PMC10984071 DOI: 10.1101/2024.03.14.24304298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Objective To identify imaging subtypes of the cortico-basal syndrome (CBS) based solely on a data-driven assessment of MRI atrophy patterns, and investigate whether these subtypes provide information on the underlying pathology. Methods We applied Subtype and Stage Inference (SuStaIn), a machine learning algorithm that identifies groups of individuals with distinct biomarker progression patterns, to a large cohort of 135 CBS cases (52 had a pathological or biomarker defined diagnosis) and 252 controls. The model was fit using volumetric features extracted from baseline T1-weighted MRI scans and validated using follow-up MRI. We compared the clinical phenotypes of each subtype and investigated whether there were differences in associated pathology between the subtypes. Results SuStaIn identified two subtypes with distinct sequences of atrophy progression; four-repeat-tauopathy confirmed cases were most commonly assigned to the Subcortical subtype (83% of CBS-PSP and 75% of CBS-CBD), while CBS-AD was most commonly assigned to the Fronto-parieto-occipital subtype (81% of CBS-AD). Subtype assignment was stable at follow-up (98% of cases), and individuals consistently progressed to higher stages (100% stayed at the same stage or progressed), supporting the model's ability to stage progression. Interpretation By jointly modelling disease stage and subtype, we provide data-driven evidence for at least two distinct and longitudinally stable spatiotemporal subtypes of atrophy in CBS that are associated with different underlying pathologies. In the absence of sensitive and specific biomarkers, accurately subtyping and staging individuals with CBS at baseline has important implications for screening on entry into clinical trials, as well as for tracking disease progression.
Collapse
Affiliation(s)
- W J Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - C Shand
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - E G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - M Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London, UK
- Centre for Cognitive and Clinical Neuroscience, Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, London, UK
| | - D M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - L VandeVrede
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA, USA
| | - H W Heuer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA, USA
| | - A L Young
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - N Oxtoby
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - D C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - J B Rowe
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge UK
| | - H R Morris
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK
- Movement Disorders Centre, University College London Queen Square Institute of Neurology, London, UK
| | - A L Boxer
- Centre for Cognitive and Clinical Neuroscience, Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, London, UK
| | - J D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - P A Wijeratne
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Informatics, University of Sussex, Brighton, BN1 9RH, UK
| |
Collapse
|
13
|
Haut F, Argyrousi EK, Arancio O. Re-Arranging the Puzzle between the Amyloid-Beta and Tau Pathology: An APP-Centric Approach. Int J Mol Sci 2023; 25:259. [PMID: 38203429 PMCID: PMC10779219 DOI: 10.3390/ijms25010259] [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: 09/21/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
After several years of research in the field of Alzheimer's disease (AD), it is still unclear how amyloid-beta (Aβ) and Tau, two key hallmarks of the disease, mediate the neuropathogenic events that lead to AD. Current data challenge the "Amyloid Cascade Hypothesis" that has prevailed in the field of AD, stating that Aβ precedes and triggers Tau pathology that will eventually become the toxic entity in the progression of the disease. This perspective also led the field of therapeutic approaches towards the development of strategies that target Aβ or Tau. In the present review, we discuss recent literature regarding the neurotoxic role of both Aβ and Tau in AD, as well as their physiological function in the healthy brain. Consequently, we present studies suggesting that Aβ and Tau act independently of each other in mediating neurotoxicity in AD, thereafter, re-evaluating the "Amyloid Cascade Hypothesis" that places Tau pathology downstream of Aβ. More recent studies have confirmed that both Aβ and Tau could propagate the disease and induce synaptic and memory impairments via the amyloid precursor protein (APP). This finding is not only interesting from a mechanistic point of view since it provides better insights into the AD pathogenesis but also from a therapeutic point of view since it renders APP a common downstream effector for both Aβ and Tau. Subsequently, therapeutic strategies that act on APP might provide a more viable and physiologically relevant approach for targeting AD.
Collapse
Affiliation(s)
- Florence Haut
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S, New York, NY 10032, USA; (F.H.); (E.K.A.)
| | - Elentina K. Argyrousi
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S, New York, NY 10032, USA; (F.H.); (E.K.A.)
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S, New York, NY 10032, USA; (F.H.); (E.K.A.)
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
14
|
Congdon EE, Ji C, Tetlow AM, Jiang Y, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease: current status and future directions. Nat Rev Neurol 2023; 19:715-736. [PMID: 37875627 PMCID: PMC10965012 DOI: 10.1038/s41582-023-00883-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Alzheimer disease (AD) is the most common cause of dementia in older individuals. AD is characterized pathologically by amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the brain, with associated loss of synapses and neurons, which eventually results in dementia. Many of the early attempts to develop treatments for AD focused on Aβ, but a lack of efficacy of these treatments in terms of slowing disease progression led to a change of strategy towards targeting of tau pathology. Given that tau shows a stronger correlation with symptom severity than does Aβ, targeting of tau is more likely to be efficacious once cognitive decline begins. Anti-tau therapies initially focused on post-translational modifications, inhibition of tau aggregation and stabilization of microtubules. However, trials of many potential drugs were discontinued because of toxicity and/or lack of efficacy. Currently, the majority of tau-targeting agents in clinical trials are immunotherapies. In this Review, we provide an update on the results from the initial immunotherapy trials and an overview of new therapeutic candidates that are in clinical development, as well as considering future directions for tau-targeting therapies.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Changyi Ji
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Amber M Tetlow
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
15
|
Mangiafico SP, Tuo QZ, Li XL, Liu Y, Haralambous C, Ding XL, Ayton S, Wang Q, Laybutt DR, Chan JY, Zhang X, Kos C, Thomas HE, Loudovaris T, Yang CH, Joannides CN, Lamont BJ, Dai L, He HH, Dong B, Andrikopoulos S, Bush AI, Lei P. Tau suppresses microtubule-regulated pancreatic insulin secretion. Mol Psychiatry 2023; 28:3982-3993. [PMID: 37735502 DOI: 10.1038/s41380-023-02267-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023]
Abstract
Tau protein is implicated in the pathogenesis of Alzheimer's disease (AD) and other tauopathies, but its physiological function is in debate. Mostly explored in the brain, tau is also expressed in the pancreas. We further explored the mechanism of tau's involvement in the regulation of glucose-stimulated insulin secretion (GSIS) in islet β-cells, and established a potential relationship between type 2 diabetes mellitus (T2DM) and AD. We demonstrate that pancreatic tau is crucial for insulin secretion regulation and glucose homeostasis. Tau levels were found to be elevated in β-islet cells of patients with T2DM, and loss of tau enhanced insulin secretion in cell lines, drosophila, and mice. Pharmacological or genetic suppression of tau in the db/db diabetic mouse model normalized glucose levels by promoting insulin secretion and was recapitulated by pharmacological inhibition of microtubule assembly. Clinical studies further showed that serum tau protein was positively correlated with blood glucose levels in healthy controls, which was lost in AD. These findings present tau as a common therapeutic target between AD and T2DM.
Collapse
Affiliation(s)
- Salvatore P Mangiafico
- Department of Medicine, Austin Hospital, University of Melbourne, Heidelberg, VIC, 3084, Australia
| | - Qing-Zhang Tuo
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Xiao-Lan Li
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Yu Liu
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Christian Haralambous
- Department of Medicine, Austin Hospital, University of Melbourne, Heidelberg, VIC, 3084, Australia
| | - Xu-Long Ding
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Qing Wang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - D Ross Laybutt
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, 2010, Australia
| | - Jeng Yie Chan
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, 2010, Australia
| | - Xiang Zhang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Cameron Kos
- St. Vincent's Institute of Medical Research and Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research and Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Thomas Loudovaris
- St. Vincent's Institute of Medical Research and Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, 3065, Australia
- Institute for Cellular Transplantation, Department of Surgery, College of Medicine, University of Arizona, Tucson, AZ, 85724-5066, USA
| | - Chieh-Hsin Yang
- Department of Medicine, Austin Hospital, University of Melbourne, Heidelberg, VIC, 3084, Australia
| | - Christos N Joannides
- Department of Medicine, Austin Hospital, University of Melbourne, Heidelberg, VIC, 3084, Australia
| | - Benjamin J Lamont
- Department of Medicine, Austin Hospital, University of Melbourne, Heidelberg, VIC, 3084, Australia
| | - Lunzhi Dai
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Hai-Huai He
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Biao Dong
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Sofianos Andrikopoulos
- Department of Medicine, Austin Hospital, University of Melbourne, Heidelberg, VIC, 3084, Australia.
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
16
|
Grossman M, Seeley WW, Boxer AL, Hillis AE, Knopman DS, Ljubenov PA, Miller B, Piguet O, Rademakers R, Whitwell JL, Zetterberg H, van Swieten JC. Frontotemporal lobar degeneration. Nat Rev Dis Primers 2023; 9:40. [PMID: 37563165 DOI: 10.1038/s41572-023-00447-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 08/12/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is one of the most common causes of early-onset dementia and presents with early social-emotional-behavioural and/or language changes that can be accompanied by a pyramidal or extrapyramidal motor disorder. About 20-25% of individuals with FTLD are estimated to carry a mutation associated with a specific FTLD pathology. The discovery of these mutations has led to important advances in potentially disease-modifying treatments that aim to slow progression or delay disease onset and has improved understanding of brain functioning. In both mutation carriers and those with sporadic disease, the most common underlying diagnoses are linked to neuronal and glial inclusions containing tau (FTLD-tau) or TDP-43 (FTLD-TDP), although 5-10% of patients may have inclusions containing proteins from the FUS-Ewing sarcoma-TAF15 family (FTLD-FET). Biomarkers definitively identifying specific pathological entities in sporadic disease have been elusive, which has impeded development of disease-modifying treatments. Nevertheless, disease-monitoring biofluid and imaging biomarkers are becoming increasingly sophisticated and are likely to serve as useful measures of treatment response during trials of disease-modifying treatments. Symptomatic trials using novel approaches such as transcranial direct current stimulation are also beginning to show promise.
Collapse
Affiliation(s)
- Murray Grossman
- Department of Neurology and Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - William W Seeley
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| | - Adam L Boxer
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Argye E Hillis
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Peter A Ljubenov
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce Miller
- Departments of Neurology and Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Olivier Piguet
- School of Psychology and Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
| | - Rosa Rademakers
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The University of Gothenburg, Mölndal, Sweden
- Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | | |
Collapse
|
17
|
Peng Y, Jin H, Xue YH, Chen Q, Yao SY, Du MQ, Liu S. Current and future therapeutic strategies for Alzheimer's disease: an overview of drug development bottlenecks. Front Aging Neurosci 2023; 15:1206572. [PMID: 37600514 PMCID: PMC10438465 DOI: 10.3389/fnagi.2023.1206572] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Alzheimer's disease (AD) is the most common chronic neurodegenerative disease worldwide. It causes cognitive dysfunction, such as aphasia and agnosia, and mental symptoms, such as behavioral abnormalities; all of which place a significant psychological and economic burden on the patients' families. No specific drugs are currently available for the treatment of AD, and the current drugs for AD only delay disease onset and progression. The pathophysiological basis of AD involves abnormal deposition of beta-amyloid protein (Aβ), abnormal tau protein phosphorylation, decreased activity of acetylcholine content, glutamate toxicity, autophagy, inflammatory reactions, mitochondria-targeting, and multi-targets. The US Food and Drug Administration (FDA) has approved five drugs for clinical use: tacrine, donepezil, carbalatine, galantamine, memantine, and lecanemab. We have focused on the newer drugs that have undergone clinical trials, most of which have not been successful as a result of excessive clinical side effects or poor efficacy. Although aducanumab received rapid approval from the FDA on 7 June 2021, its long-term safety and tolerability require further monitoring and confirmation. In this literature review, we aimed to explore the possible pathophysiological mechanisms underlying the occurrence and development of AD. We focused on anti-Aβ and anti-tau drugs, mitochondria-targeting and multi-targets, commercially available drugs, bottlenecks encountered in drug development, and the possible targets and therapeutic strategies for future drug development. We hope to present new concepts and methods for future drug therapies for AD.
Collapse
Affiliation(s)
- Yong Peng
- Neurology Department, The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- Neurology Department, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Hong Jin
- Neurology Department, The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- Neurology Department, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Ya-hui Xue
- Neurology Department, The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- Neurology Department, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Quan Chen
- Neurology Department, The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- Neurology Department, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Shun-yu Yao
- Neurology Department, The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- Neurology Department, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Miao-qiao Du
- Neurology Department, The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- Neurology Department, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Shu Liu
- Neurology Department, The First Affiliated Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
- Neurology Department, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| |
Collapse
|
18
|
Neylan KD, Miller BL. New Approaches to the Treatment of Frontotemporal Dementia. Neurotherapeutics 2023; 20:1055-1065. [PMID: 37157041 PMCID: PMC10457270 DOI: 10.1007/s13311-023-01380-6] [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] [Accepted: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Frontotemporal dementia (FTD) comprises a diverse group of clinical neurodegenerative syndromes characterized by progressive changes in behavior, personality, executive function, language, and motor function. Approximately 20% of FTD cases have a known genetic cause. The three most common genetic mutations causing FTD are discussed. Frontotemporal lobar degeneration refers to the heterogeneous group of neuropathology underlying FTD clinical syndromes. While there are no current disease-modifying treatments for FTD, management includes off-label pharmacotherapy and non-pharmacological approaches to target symptoms. The utility of several different drug classes is discussed. Medications used in the treatment of Alzheimer's disease have no benefit in FTD and can worsen neuropsychiatric symptoms. Non-pharmacological approaches to management include lifestyle modifications, speech-, occupational-, and physical therapy, peer and caregiver support, and safety considerations. Recent developments in the understanding of the genetics, pathophysiology, neuropathology, and neuroimmunology underlying FTD clinical syndromes have expanded possibilities for disease-modifying and symptom-targeted treatments. Different pathogenetic mechanisms are targeted in several active clinical trials, opening up exciting possibilities for breakthrough advances in treatment and management of FTD spectrum disorders.
Collapse
Affiliation(s)
- Kyra D Neylan
- University of California San Francisco Memory and Aging Center, San Francisco, USA.
| | - Bruce L Miller
- University of California San Francisco Memory and Aging Center, San Francisco, USA
| |
Collapse
|
19
|
Samudra N, Lane-Donovan C, VandeVrede L, Boxer AL. Tau pathology in neurodegenerative disease: disease mechanisms and therapeutic avenues. J Clin Invest 2023; 133:e168553. [PMID: 37317972 PMCID: PMC10266783 DOI: 10.1172/jci168553] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023] Open
Abstract
Tauopathies are disorders associated with tau protein dysfunction and insoluble tau accumulation in the brain at autopsy. Multiple lines of evidence from human disease, as well as nonclinical translational models, suggest that tau has a central pathologic role in these disorders, historically thought to be primarily related to tau gain of toxic function. However, a number of tau-targeting therapies with various mechanisms of action have shown little promise in clinical trials in different tauopathies. We review what is known about tau biology, genetics, and therapeutic mechanisms that have been tested in clinical trials to date. We discuss possible reasons for failures of these therapies, such as use of imperfect nonclinical models that do not predict human effects for drug development; heterogeneity of human tau pathologies which may lead to variable responses to therapy; and ineffective therapeutic mechanisms, such as targeting of the wrong tau species or protein epitope. Innovative approaches to human clinical trials can help address some of the difficulties that have plagued our field's development of tau-targeting therapies thus far. Despite limited clinical success to date, as we continue to refine our understanding of tau's pathogenic mechanism(s) in different neurodegenerative diseases, we remain optimistic that tau-targeting therapies will eventually play a central role in the treatment of tauopathies.
Collapse
|
20
|
Scotton WJ, Shand C, Todd E, Bocchetta M, Cash DM, VandeVrede L, Heuer H, Young AL, Oxtoby N, Alexander DC, Rowe JB, Morris HR, Boxer AL, Rohrer JD, Wijeratne PA. Uncovering spatiotemporal patterns of atrophy in progressive supranuclear palsy using unsupervised machine learning. Brain Commun 2023; 5:fcad048. [PMID: 36938523 PMCID: PMC10016410 DOI: 10.1093/braincomms/fcad048] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/22/2022] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
To better understand the pathological and phenotypic heterogeneity of progressive supranuclear palsy and the links between the two, we applied a novel unsupervised machine learning algorithm (Subtype and Stage Inference) to the largest MRI data set to date of people with clinically diagnosed progressive supranuclear palsy (including progressive supranuclear palsy-Richardson and variant progressive supranuclear palsy syndromes). Our cohort is comprised of 426 progressive supranuclear palsy cases, of which 367 had at least one follow-up scan, and 290 controls. Of the progressive supranuclear palsy cases, 357 were clinically diagnosed with progressive supranuclear palsy-Richardson, 52 with a progressive supranuclear palsy-cortical variant (progressive supranuclear palsy-frontal, progressive supranuclear palsy-speech/language, or progressive supranuclear palsy-corticobasal), and 17 with a progressive supranuclear palsy-subcortical variant (progressive supranuclear palsy-parkinsonism or progressive supranuclear palsy-progressive gait freezing). Subtype and Stage Inference was applied to volumetric MRI features extracted from baseline structural (T1-weighted) MRI scans and then used to subtype and stage follow-up scans. The subtypes and stages at follow-up were used to validate the longitudinal consistency of subtype and stage assignments. We further compared the clinical phenotypes of each subtype to gain insight into the relationship between progressive supranuclear palsy pathology, atrophy patterns, and clinical presentation. The data supported two subtypes, each with a distinct progression of atrophy: a 'subcortical' subtype, in which early atrophy was most prominent in the brainstem, ventral diencephalon, superior cerebellar peduncles, and the dentate nucleus, and a 'cortical' subtype, in which there was early atrophy in the frontal lobes and the insula alongside brainstem atrophy. There was a strong association between clinical diagnosis and the Subtype and Stage Inference subtype with 82% of progressive supranuclear palsy-subcortical cases and 81% of progressive supranuclear palsy-Richardson cases assigned to the subcortical subtype and 82% of progressive supranuclear palsy-cortical cases assigned to the cortical subtype. The increasing stage was associated with worsening clinical scores, whilst the 'subcortical' subtype was associated with worse clinical severity scores compared to the 'cortical subtype' (progressive supranuclear palsy rating scale and Unified Parkinson's Disease Rating Scale). Validation experiments showed that subtype assignment was longitudinally stable (95% of scans were assigned to the same subtype at follow-up) and individual staging was longitudinally consistent with 90% remaining at the same stage or progressing to a later stage at follow-up. In summary, we applied Subtype and Stage Inference to structural MRI data and empirically identified two distinct subtypes of spatiotemporal atrophy in progressive supranuclear palsy. These image-based subtypes were differentially enriched for progressive supranuclear palsy clinical syndromes and showed different clinical characteristics. Being able to accurately subtype and stage progressive supranuclear palsy patients at baseline has important implications for screening patients on entry to clinical trials, as well as tracking disease progression.
Collapse
Affiliation(s)
- William J Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Cameron Shand
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| | - Emily Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Lawren VandeVrede
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA 94158, USA
| | - Hilary Heuer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA 94158, USA
| | - Alexandra L Young
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Neil Oxtoby
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| | - James B Rowe
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Medical Research Council Cognition and Brain Sciences Unit, Cambridge CB2 0QQ, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- Movement Disorders Centre, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA 94158, USA
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Peter A Wijeratne
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1V 6LJ, UK
| |
Collapse
|
21
|
Duque KR, Vizcarra JA, Hill EJ, Espay AJ. Disease-modifying vs symptomatic treatments: Splitting over lumping. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:187-209. [PMID: 36803811 DOI: 10.1016/b978-0-323-85555-6.00020-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Clinical trials of putative disease-modifying therapies in neurodegeneration have obeyed the century-old principle of convergence, or lumping, whereby any feature of a clinicopathologic disease entity is considered relevant to most of those affected. While this convergent approach has resulted in important successes in trials of symptomatic therapies, largely aimed at correcting common neurotransmitter deficiencies (e.g., cholinergic deficiency in Alzheimer's disease or dopaminergic deficiency in Parkinson's disease), it has been consistently futile in trials of neuroprotective or disease-modifying interventions. As individuals affected by the same neurodegenerative disorder do not share the same biological drivers, splitting such disease into small molecular/biological subtypes, to match people to therapies most likely to benefit them, is vital in the pursuit of disease modification. We here discuss three paths toward the splitting needed for future successes in precision medicine: (1) encourage the development of aging cohorts agnostic to phenotype in order to enact a biology-to-phenotype direction of biomarker development and validate divergence biomarkers (present in some, absent in most); (2) demand bioassay-based recruitment of subjects into disease-modifying trials of putative neuroprotective interventions in order to match the right therapies to the right recipients; and (3) evaluate promising epidemiologic leads of presumed pathogenetic potential using Mendelian randomization studies before designing the corresponding clinical trials. The reconfiguration of disease-modifying efforts for patients with neurodegenerative disorders will require a paradigm shift from lumping to splitting and from proteinopathy to proteinopenia.
Collapse
Affiliation(s)
- Kevin R Duque
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Joaquin A Vizcarra
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Emily J Hill
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States.
| |
Collapse
|
22
|
Coughlin DG, Litvan I. Investigational therapeutics for the treatment of progressive supranuclear palsy. Expert Opin Investig Drugs 2022; 31:813-823. [DOI: 10.1080/13543784.2022.2087179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- David G Coughlin
- Department of Neurosciences, University of California San Diego, San Diego, 92093, CA
| | - Irene Litvan
- Department of Neurosciences, University of California San Diego, San Diego, 92093, CA
| |
Collapse
|
23
|
Scotton WJ, Bocchetta M, Todd E, Cash DM, Oxtoby N, VandeVrede L, Heuer H, Alexander DC, Rowe JB, Morris HR, Boxer A, Rohrer JD, Wijeratne PA. A data-driven model of brain volume changes in progressive supranuclear palsy. Brain Commun 2022; 4:fcac098. [PMID: 35602649 PMCID: PMC9118104 DOI: 10.1093/braincomms/fcac098] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/08/2021] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
The most common clinical phenotype of progressive supranuclear palsy is Richardson syndrome, characterized by levodopa unresponsive symmetric parkinsonism, with a vertical supranuclear gaze palsy, early falls and cognitive impairment. There is currently no detailed understanding of the full sequence of disease pathophysiology in progressive supranuclear palsy. Determining the sequence of brain atrophy in progressive supranuclear palsy could provide important insights into the mechanisms of disease progression, as well as guide patient stratification and monitoring for clinical trials. We used a probabilistic event-based model applied to cross-sectional structural MRI scans in a large international cohort, to determine the sequence of brain atrophy in clinically diagnosed progressive supranuclear palsy Richardson syndrome. A total of 341 people with Richardson syndrome (of whom 255 had 12-month follow-up imaging) and 260 controls were included in the study. We used a combination of 12-month follow-up MRI scans, and a validated clinical rating score (progressive supranuclear palsy rating scale) to demonstrate the longitudinal consistency and utility of the event-based model's staging system. The event-based model estimated that the earliest atrophy occurs in the brainstem and subcortical regions followed by progression caudally into the superior cerebellar peduncle and deep cerebellar nuclei, and rostrally to the cortex. The sequence of cortical atrophy progresses in an anterior to posterior direction, beginning in the insula and then the frontal lobe before spreading to the temporal, parietal and finally the occipital lobe. This in vivo ordering accords with the post-mortem neuropathological staging of progressive supranuclear palsy and was robust under cross-validation. Using longitudinal information from 12-month follow-up scans, we demonstrate that subjects consistently move to later stages over this time interval, supporting the validity of the model. In addition, both clinical severity (progressive supranuclear palsy rating scale) and disease duration were significantly correlated with the predicted subject event-based model stage (P < 0.01). Our results provide new insights into the sequence of atrophy progression in progressive supranuclear palsy and offer potential utility to stratify people with this disease on entry into clinical trials based on disease stage, as well as track disease progression.
Collapse
Affiliation(s)
- W. J. Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen
Square Institute of Neurology, University College London, London, UK
| | - M. Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen
Square Institute of Neurology, University College London, London, UK
| | - E. Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen
Square Institute of Neurology, University College London, London, UK
| | - D. M. Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen
Square Institute of Neurology, University College London, London, UK
| | - N. Oxtoby
- Centre for Medical Image Computing, Department of Computer Science, University
College London, London, UK
| | - L. VandeVrede
- Department of Neurology, Memory and Aging Center, University of
California, San Francisco, CA, USA
| | - H. Heuer
- Department of Neurology, Memory and Aging Center, University of
California, San Francisco, CA, USA
| | | | - D. C. Alexander
- Centre for Medical Image Computing, Department of Computer Science, University
College London, London, UK
| | - J. B. Rowe
- Department of Clinical Neurosciences, Cambridge University, Cambridge
University Hospitals NHS Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge
University, Cambridge, UK
| | - H. R. Morris
- Department of Clinical and Movement Neurosciences, University College London
Queen Square Institute of Neurology, London, UK
- Movement Disorders Centre, University College London Queen Square Institute of
Neurology, London, UK
| | - A. Boxer
- Department of Neurology, Memory and Aging Center, University of
California, San Francisco, CA, USA
| | - J. D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen
Square Institute of Neurology, University College London, London, UK
| | - P. A. Wijeratne
- Centre for Medical Image Computing, Department of Computer Science, University
College London, London, UK
| |
Collapse
|
24
|
Kassab A, Rizk N, Prakash S. The Role of Systemic Filtrating Organs in Aging and Their Potential in Rejuvenation Strategies. Int J Mol Sci 2022; 23:ijms23084338. [PMID: 35457154 PMCID: PMC9025381 DOI: 10.3390/ijms23084338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 11/26/2022] Open
Abstract
Advances in aging studies brought about by heterochronic parabiosis suggest that aging might be a reversable process that is affected by changes in the systemic milieu of organs and cells. Given the broadness of such a systemic approach, research to date has mainly questioned the involvement of “shared organs” versus “circulating factors”. However, in the absence of a clear understanding of the chronological development of aging and a unified platform to evaluate the successes claimed by specific rejuvenation methods, current literature on this topic remains scattered. Herein, aging is assessed from an engineering standpoint to isolate possible aging potentiators via a juxtaposition between biological and mechanical systems. Such a simplification provides a general framework for future research in the field and examines the involvement of various factors in aging. Based on this simplified overview, the kidney as a filtration organ is clearly implicated, for the first time, with the aging phenomenon, necessitating a re-evaluation of current rejuvenation studies to untangle the extent of its involvement and its possible role as a potentiator in aging. Based on these findings, the review concludes with potential translatable and long-term therapeutics for aging while offering a critical view of rejuvenation methods proposed to date.
Collapse
Affiliation(s)
- Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC H3A 2BA, Canada
| | - Nasser Rizk
- Department of Biomedical Sciences, College of Health Sciences-QU-Health, Qatar University, Doha 2713, Qatar
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC H3A 2BA, Canada
| |
Collapse
|
25
|
Zhang Y, Wu KM, Yang L, Dong Q, Yu JT. Tauopathies: new perspectives and challenges. Mol Neurodegener 2022; 17:28. [PMID: 35392986 PMCID: PMC8991707 DOI: 10.1186/s13024-022-00533-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tauopathies are a class of neurodegenerative disorders characterized by neuronal and/or glial tau-positive inclusions. MAIN BODY Clinically, tauopathies can present with a range of phenotypes that include cognitive/behavioral-disorders, movement disorders, language disorders and non-specific amnestic symptoms in advanced age. Pathologically, tauopathies can be classified based on the predominant tau isoforms that are present in the inclusion bodies (i.e., 3R, 4R or equal 3R:4R ratio). Imaging, cerebrospinal fluid (CSF) and blood-based tau biomarkers have the potential to be used as a routine diagnostic strategy and in the evaluation of patients with tauopathies. As tauopathies are strongly linked neuropathologically and genetically to tau protein abnormalities, there is a growing interest in pursuing of tau-directed therapeutics for the disorders. Here we synthesize emerging lessons on tauopathies from clinical, pathological, genetic, and experimental studies toward a unified concept of these disorders that may accelerate the therapeutics. CONCLUSIONS Since tauopathies are still untreatable diseases, efforts have been made to depict clinical and pathological characteristics, identify biomarkers, elucidate underlying pathogenesis to achieve early diagnosis and develop disease-modifying therapies.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Liu Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| |
Collapse
|
26
|
Islam M, Shen F, Regmi D, Du D. Therapeutic strategies for tauopathies and drug repurposing as a potential approach. Biochem Pharmacol 2022; 198:114979. [PMID: 35219701 PMCID: PMC9159505 DOI: 10.1016/j.bcp.2022.114979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/03/2022] [Accepted: 02/21/2022] [Indexed: 11/26/2022]
Abstract
Tauopathies are neurodegenerative diseases characterized by the deposition of abnormal tau in the brain. To date, there are no disease-modifying therapies approved by the U.S. Food and Drug Administration (US FDA) for the treatment of tauopathies. In the past decades, extensive efforts have been provided to develop disease-modifying therapies to treat tauopathies. Specifically, exploring existing drugs with the intent of repurposing for the treatment of tauopathies affords a reasonable alternative to discover potent drugs for treating these formidable diseases. Drug repurposing will not only reduce formulation and development stage effort and cost but will also take a key advantage of the established toxicological studies, which is one of the main causes of clinical trial failure of new molecules. In this review, we provide an overview of the current treatment strategies for tauopathies and the recent progress in drug repurposing as an alternative approach to treat tauopathies.
Collapse
Affiliation(s)
- Majedul Islam
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States.
| | - Fengyun Shen
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Deepika Regmi
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Deguo Du
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States.
| |
Collapse
|
27
|
Saito Y, Kamagata K, Wijeratne PA, Andica C, Uchida W, Takabayashi K, Fujita S, Akashi T, Wada A, Shimoji K, Hori M, Masutani Y, Alexander DC, Aoki S. Temporal Progression Patterns of Brain Atrophy in Corticobasal Syndrome and Progressive Supranuclear Palsy Revealed by Subtype and Stage Inference (SuStaIn). Front Neurol 2022; 13:814768. [PMID: 35280291 PMCID: PMC8914081 DOI: 10.3389/fneur.2022.814768] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Differentiating corticobasal degeneration presenting with corticobasal syndrome (CBD-CBS) from progressive supranuclear palsy with Richardson's syndrome (PSP-RS), particularly in early stages, is often challenging because the neurodegenerative conditions closely overlap in terms of clinical presentation and pathology. Although volumetry using brain magnetic resonance imaging (MRI) has been studied in patients with CBS and PSP-RS, studies assessing the progression of brain atrophy are limited. Therefore, we aimed to reveal the difference in the temporal progression patterns of brain atrophy between patients with CBS and those with PSP-RS purely based on cross-sectional data using Subtype and Stage Inference (SuStaIn)—a novel, unsupervised machine learning technique that integrates clustering and disease progression modeling. We applied SuStaIn to the cross-sectional regional brain volumes of 25 patients with CBS, 39 patients with typical PSP-RS, and 50 healthy controls to estimate the two disease subtypes and trajectories of CBS and PSP-RS, which have distinct atrophy patterns. The progression model and classification accuracy of CBS and PSP-RS were compared with those of previous studies to evaluate the performance of SuStaIn. SuStaIn identified distinct temporal progression patterns of brain atrophy for CBS and PSP-RS, which were largely consistent with previous evidence, with high reproducibility (99.7%) under cross-validation. We classified these diseases with high accuracy (0.875) and sensitivity (0.680 and 1.000, respectively) based on cross-sectional structural brain MRI data; the accuracy was higher than that reported in previous studies. Moreover, SuStaIn stage correctly reflected disease severity without the label of disease stage, such as disease duration. Furthermore, SuStaIn also showed the genialized performance of differentiation and reflection for CBS and PSP-RS. Thus, SuStaIn has potential for improving our understanding of disease mechanisms, accurately stratifying patients, and providing prognoses for patients with CBS and PSP-RS.
Collapse
Affiliation(s)
- Yuya Saito
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- *Correspondence: Koji Kamagata
| | - Peter A. Wijeratne
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Christina Andica
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Wataru Uchida
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kaito Takabayashi
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shohei Fujita
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Radiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Toshiaki Akashi
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihiko Wada
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keigo Shimoji
- Department of Radiology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Masaaki Hori
- Department of Radiology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yoshitaka Masutani
- Department of Biomedical Information Sciences, Hiroshima City University Graduate School of Information Sciences, Hiroshima, Japan
| | - Daniel C. Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Shigeki Aoki
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Alquezar C, Schoch KM, Geier EG, Ramos EM, Scrivo A, Li KH, Argouarch AR, Mlynarski EE, Dombroski B, DeTure M, Dickson DW, Yokoyama JS, Cuervo AM, Burlingame AL, Schellenberg GD, Miller TM, Miller BL, Kao AW. TSC1 loss increases risk for tauopathy by inducing tau acetylation and preventing tau clearance via chaperone-mediated autophagy. SCIENCE ADVANCES 2021; 7:eabg3897. [PMID: 34739309 PMCID: PMC8570595 DOI: 10.1126/sciadv.abg3897] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 09/17/2021] [Indexed: 05/20/2023]
Abstract
Age-associated neurodegenerative disorders demonstrating tau-laden intracellular inclusions are known as tauopathies. We previously linked a loss-of-function mutation in the TSC1 gene to tau accumulation and frontotemporal lobar degeneration. Now, we have identified genetic variants in TSC1 that decrease TSC1/hamartin levels and predispose to tauopathies such as Alzheimer’s disease and progressive supranuclear palsy. Cellular and murine models of TSC1 haploinsufficiency, as well as human brains carrying a TSC1 risk variant, accumulated tau protein that exhibited aberrant acetylation. This acetylation hindered tau degradation via chaperone-mediated autophagy, thereby leading to its accumulation. Aberrant tau acetylation in TSC1 haploinsufficiency resulted from the dysregulation of both p300 acetyltransferase and SIRT1 deacetylase. Pharmacological modulation of either enzyme restored tau levels. This study substantiates TSC1 as a novel tauopathy risk gene and includes TSC1 haploinsufficiency as a genetic model for tauopathies. In addition, these findings promote tau acetylation as a rational target for tauopathy therapeutics and diagnostic.
Collapse
Affiliation(s)
- Carolina Alquezar
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kathleen M. Schoch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ethan G. Geier
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eliana Marisa Ramos
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Aurora Scrivo
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Kathy H. Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Andrea R. Argouarch
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Elisabeth E. Mlynarski
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Beth Dombroski
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Michael DeTure
- Department of Neuroscience, The Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Dennis W. Dickson
- Department of Neuroscience, The Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Jennifer S. Yokoyama
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ana M. Cuervo
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Alma L. Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Timothy M. Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bruce L. Miller
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Aimee W. Kao
- UCSF Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
- Corresponding author.
| |
Collapse
|
29
|
[Neuroprotective treatment of tauopathies]. DER NERVENARZT 2021; 92:1227-1238. [PMID: 34652482 DOI: 10.1007/s00115-021-01210-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Tau pathology is now considered to be the main cause of a wide spectrum of neurodegenerative diseases, which are collectively referred to as tauopathies. These include primary tauopathies, in which tau plays the main role in the pathogenesis as well as secondary tauopathies, such as Alzheimer's disease, in which amyloid beta also plays a substantial role in the disease process in addition to the tau pathology. Primary tauopathies include progressive supranuclear palsy, corticobasal degeneration, Pick's disease and rare hereditary tauopathies, which are referred to as frontotemporal lobar degeneration with microtubule-associated protein tau (MAPT) mutation. Tauopathies differ from each other pathologically by the affected brain regions and cell types as well as by the biochemical characteristics of the aggregated tau protein. Various tau-centered neuroprotective treatment approaches are currently in preclinical and clinical development. They target different mechanisms, including the reduction of tau expression, inhibition of tau aggregation, dissolution of tau aggregates, improvement of cellular mechanisms to eliminate toxic tau species, stabilization of microtubules and prevention of intercellular tau spreading. This review article gives an overview of tauopathies and the current concepts for the development of disease-modifying treatment.
Collapse
|
30
|
Stamelou M, Respondek G, Giagkou N, Whitwell JL, Kovacs GG, Höglinger GU. Evolving concepts in progressive supranuclear palsy and other 4-repeat tauopathies. Nat Rev Neurol 2021; 17:601-620. [PMID: 34426686 DOI: 10.1038/s41582-021-00541-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Tauopathies are classified according to whether tau deposits predominantly contain tau isoforms with three or four repeats of the microtubule-binding domain. Those in which four-repeat (4R) tau predominates are known as 4R-tauopathies, and include progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, globular glial tauopathies and conditions associated with specific MAPT mutations. In these diseases, 4R-tau deposits are found in various cell types and anatomical regions of the brain and the conditions share pathological, pathophysiological and clinical characteristics. Despite being considered 'prototype' tauopathies and, therefore, ideal for studying neuroprotective agents, 4R-tauopathies are still severe and untreatable diseases for which no validated biomarkers exist. However, advances in research have addressed the issues of phenotypic overlap, early clinical diagnosis, pathophysiology and identification of biomarkers, setting a road map towards development of treatments. New clinical criteria have been developed and large cohorts with early disease are being followed up in prospective studies. New clinical trial readouts are emerging and biomarker research is focused on molecular pathways that have been identified. Lessons learned from failed trials of neuroprotective drugs are being used to design new trials. In this Review, we present an overview of the latest research in 4R-tauopathies, with a focus on progressive supranuclear palsy, and discuss how current evidence dictates ongoing and future research goals.
Collapse
Affiliation(s)
- Maria Stamelou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece. .,European University of Cyprus, Nicosia, Cyprus. .,Philipps University, Marburg, Germany.
| | - Gesine Respondek
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Nikolaos Giagkou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece
| | | | - Gabor G Kovacs
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Toronto, Ontario, Canada.,Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Günter U Höglinger
- Department of Neurology, Hanover Medical School, Hanover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
31
|
Imbimbo BP, Ippati S, Watling M, Balducci C. A critical appraisal of tau-targeting therapies for primary and secondary tauopathies. Alzheimers Dement 2021; 18:1008-1037. [PMID: 34533272 DOI: 10.1002/alz.12453] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Primary tauopathies are neurological disorders in which tau protein deposition is the predominant pathological feature. Alzheimer's disease is a secondary tauopathy with tau forming hyperphosphorylated insoluble aggregates. Tau pathology can propagate from region to region in the brain, while alterations in tau processing may impair tau physiological functions. METHODS We reviewed literature on tau biology and anti-tau drugs using PubMed, meeting abstracts, and ClnicalTrials.gov. RESULTS The past 15 years have seen >30 drugs interfering with tau aggregation, processing, and accumulation reaching the clinic. Initial results with tau aggregation inhibitors and anti-tau monoclonal antibodies have not shown clinical efficacy. DISCUSSION The reasons for these clinical failures are unclear but could be linked to the clearing of physiological forms of tau by non-specific drugs. Research is now concentrating efforts on developing reliable translational animal models and selective compounds targeting specific tau epitopes, neurotoxic tau aggregates, and post-translational tau modifications.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | - Stefania Ippati
- San Raffaele Scientific Institute, San Raffaele Hospital, Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Ltd, Reading, UK
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milan, Italy
| |
Collapse
|
32
|
Przewodowska D, Marzec W, Madetko N. Novel Therapies for Parkinsonian Syndromes-Recent Progress and Future Perspectives. Front Mol Neurosci 2021; 14:720220. [PMID: 34512258 PMCID: PMC8427499 DOI: 10.3389/fnmol.2021.720220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/23/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Atypical parkinsonian syndromes are rare, fatal neurodegenerative diseases associated with abnormal protein accumulation in the brain. Examples of these syndromes include progressive supranuclear palsy, multiple system atrophy, and corticobasal degeneration. A common clinical feature in parkinsonism is a limited improvement with levodopa. So far, there are no disease-modifying treatments to address these conditions, and therapy is only limited to the alleviation of symptoms. Diagnosis is devastating for patients, as prognosis is extremely poor, and the disease tends to progress rapidly. Currently, potential causes and neuropathological mechanisms involved in these diseases are being widely investigated. Objectives: The goal of this review is to summarize recent advances and gather emerging disease-modifying therapies that could slow the progression of atypical parkinsonian syndromes. Methods: PubMed and Google Scholar databases were searched regarding novel perspectives for atypical parkinsonism treatment. The following medical subject headings were used: "atypical parkinsonian syndromes-therapy," "treatment of atypical parkinsonian syndromes," "atypical parkinsonian syndromes-clinical trial," "therapy of tauopathy," "alpha-synucleinopathy treatment," "PSP therapy/treatment," "CBD therapy/treatment," "MSA therapy/treatment," and "atypical parkinsonian syndromes-disease modifying." All search results were manually reviewed prior to inclusion in this review. Results: Neuroinflammation, mitochondrial dysfunction, microglia activation, proteasomal impairment, and oxidative stress play a role in the neurodegenerative process. Ongoing studies and clinical trials target these components in order to suppress toxic protein accumulation. Various approaches such as stem cell therapy, anti-aggregation/anti-phosphorylation agent administration, or usage of active and passive immunization appear to have promising results. Conclusion: Presently, disease-modifying strategies for atypical parkinsonian syndromes are being actively explored, with encouraging preliminary results. This leads to an assumption that developing accurate, safe, and progression-halting treatment is not far off. Nevertheless, the further investigation remains necessary.
Collapse
Affiliation(s)
- Dominika Przewodowska
- Students' Scientific Association of the Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Weronika Marzec
- Students' Scientific Association of the Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Natalia Madetko
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
33
|
Sinsky J, Pichlerova K, Hanes J. Tau Protein Interaction Partners and Their Roles in Alzheimer's Disease and Other Tauopathies. Int J Mol Sci 2021; 22:9207. [PMID: 34502116 PMCID: PMC8431036 DOI: 10.3390/ijms22179207] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Tau protein plays a critical role in the assembly, stabilization, and modulation of microtubules, which are important for the normal function of neurons and the brain. In diseased conditions, several pathological modifications of tau protein manifest. These changes lead to tau protein aggregation and the formation of paired helical filaments (PHF) and neurofibrillary tangles (NFT), which are common hallmarks of Alzheimer's disease and other tauopathies. The accumulation of PHFs and NFTs results in impairment of physiological functions, apoptosis, and neuronal loss, which is reflected as cognitive impairment, and in the late stages of the disease, leads to death. The causes of this pathological transformation of tau protein haven't been fully understood yet. In both physiological and pathological conditions, tau interacts with several proteins which maintain their proper function or can participate in their pathological modifications. Interaction partners of tau protein and associated molecular pathways can either initiate and drive the tau pathology or can act neuroprotective, by reducing pathological tau proteins or inflammation. In this review, we focus on the tau as a multifunctional protein and its known interacting partners active in regulations of different processes and the roles of these proteins in Alzheimer's disease and tauopathies.
Collapse
Affiliation(s)
| | | | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia; (J.S.); (K.P.)
| |
Collapse
|
34
|
Wang D, Huang X, Yan L, Zhou L, Yan C, Wu J, Su Z, Huang Y. The Structure Biology of Tau and Clue for Aggregation Inhibitor Design. Protein J 2021; 40:656-668. [PMID: 34401998 DOI: 10.1007/s10930-021-10017-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
Tau is a microtubule-associated protein that is mainly expressed in central and peripheral nerve systems. Tau binds to tubulin and regulates assembly and stabilization of microtubule, thus playing a critical role in neuron morphology, axon development and navigation. Tau is highly stable under normal conditions; however, there are several factors that can induce or promote aggregation of tau, forming neurofibrillary tangles. Neurofibrillary tangles are toxic to neurons, which may be related to a series of neurodegenerative diseases including Alzheimer's disease. Thus, tau is widely accepted as an important therapeutic target for neurodegenerative diseases. While the monomeric structure of tau is highly disordered, the aggregate structure of tau is formed by closed packing of β-stands. Studies on the structure of tau and the structural transition mechanism provide valuable information on the occurrence, development, and therapy of tauopathies. In this review, we summarize recent progress on the structural investigation of tau and based on which we discuss aggregation inhibitor design.
Collapse
Affiliation(s)
- Dan Wang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Xianlong Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Lu Yan
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Luoqi Zhou
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Chang Yan
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Jinhu Wu
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Zhengding Su
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Yongqi Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China. .,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| |
Collapse
|
35
|
Bluett B, Pantelyat AY, Litvan I, Ali F, Apetauerova D, Bega D, Bloom L, Bower J, Boxer AL, Dale ML, Dhall R, Duquette A, Fernandez HH, Fleisher JE, Grossman M, Howell M, Kerwin DR, Leegwater-Kim J, Lepage C, Ljubenkov PA, Mancini M, McFarland NR, Moretti P, Myrick E, Patel P, Plummer LS, Rodriguez-Porcel F, Rojas J, Sidiropoulos C, Sklerov M, Sokol LL, Tuite PJ, VandeVrede L, Wilhelm J, Wills AMA, Xie T, Golbe LI. Best Practices in the Clinical Management of Progressive Supranuclear Palsy and Corticobasal Syndrome: A Consensus Statement of the CurePSP Centers of Care. Front Neurol 2021; 12:694872. [PMID: 34276544 PMCID: PMC8284317 DOI: 10.3389/fneur.2021.694872] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/10/2021] [Indexed: 11/16/2022] Open
Abstract
Progressive supranuclear palsy (PSP) and corticobasal syndrome (CBS; the most common phenotype of corticobasal degeneration) are tauopathies with a relentless course, usually starting in the mid-60s and leading to death after an average of 7 years. There is as yet no specific or disease-modifying treatment. Clinical deficits in PSP are numerous, involve the entire neuraxis, and present as several discrete phenotypes. They center on rigidity, bradykinesia, postural instability, gait freezing, supranuclear ocular motor impairment, dysarthria, dysphagia, incontinence, sleep disorders, frontal cognitive dysfunction, and a variety of behavioral changes. CBS presents with prominent and usually asymmetric dystonia, apraxia, myoclonus, pyramidal signs, and cortical sensory loss. The symptoms and deficits of PSP and CBS are amenable to a variety of treatment strategies but most physicians, including many neurologists, are reluctant to care for patients with these conditions because of unfamiliarity with their multiplicity of interacting symptoms and deficits. CurePSP, the organization devoted to support, research, and education for PSP and CBS, created its CurePSP Centers of Care network in North America in 2017 to improve patient access to clinical expertise and develop collaborations. The directors of the 25 centers have created this consensus document outlining best practices in the management of PSP and CBS. They formed a writing committee for each of 12 sub-topics. A 4-member Steering Committee collated and edited the contributions. The result was returned to the entire cohort of authors for further comments, which were considered for incorporation by the Steering Committee. The authors hope that this publication will serve as a convenient guide for all clinicians caring for patients with PSP and CBS and that it will improve care for patients with these devastating but manageable disorders.
Collapse
Affiliation(s)
- Brent Bluett
- Neurology, Pacific Central Coast Health Center, Dignity Health, San Luis Obispo, CA, United States
- Neurology, Stanford University, Stanford, CA, United States
| | - Alexander Y. Pantelyat
- Neurology, The Johns Hopkins Hospital, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Irene Litvan
- Neurology, University of California, San Diego, San Diego, CA, United States
| | - Farwa Ali
- Neurology, Mayo Clinic, Rochester, MN, United States
| | - Diana Apetauerova
- Neurology, Lahey Hospital and Medical Center, Burlington, MA, United States
| | - Danny Bega
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lisa Bloom
- Neurology, Surgery, University of Chicago, Chicago, IL, United States
| | - James Bower
- Neurology, Mayo Clinic, Rochester, MN, United States
| | - Adam L. Boxer
- Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Marian L. Dale
- Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Rohit Dhall
- Neurology, University of Arkansas for Medical Sciences, Little Rock, AK, United States
| | - Antoine Duquette
- Service de Neurologie, Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Service de Neurologie, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Hubert H. Fernandez
- Center for Neurological Restoration, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jori E. Fleisher
- Neurological Sciences, Rush Medical College, Rush University, Chicago, IL, United States
| | - Murray Grossman
- Neurology, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael Howell
- Neurology, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Diana R. Kerwin
- Geriatrics, Presbyterian Hospital of Dallas, Dallas, TX, United States
| | | | - Christiane Lepage
- Service de Neurologie, Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Service de Neurologie, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | | | - Martina Mancini
- Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Nikolaus R. McFarland
- Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Paolo Moretti
- Neurology, The University of Utah, Salt Lake City, UT, United States
| | - Erica Myrick
- Neurological Sciences, Rush Medical College, Rush University, Chicago, IL, United States
| | - Pritika Patel
- Neurology, Lahey Hospital and Medical Center, Burlington, MA, United States
| | - Laura S. Plummer
- Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | | | - Julio Rojas
- Neurology, University of California, San Francisco, San Francisco, CA, United States
| | | | - Miriam Sklerov
- Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Leonard L. Sokol
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Paul J. Tuite
- Neurology, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Lawren VandeVrede
- Neurology, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jennifer Wilhelm
- Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Anne-Marie A. Wills
- Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Tao Xie
- Neurology, Surgery, University of Chicago, Chicago, IL, United States
| | - Lawrence I. Golbe
- Neurology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
36
|
Giunta M, Solje E, Gardoni F, Borroni B, Benussi A. Experimental Disease-Modifying Agents for Frontotemporal Lobar Degeneration. J Exp Pharmacol 2021; 13:359-376. [PMID: 33790662 PMCID: PMC8005747 DOI: 10.2147/jep.s262352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia is a clinically, genetically and pathologically heterogeneous neurodegenerative disorder, enclosing a wide range of different pathological entities, associated with the accumulation of proteins such as tau and TPD-43. Characterized by a high hereditability, mutations in three main genes, MAPT, GRN and C9orf72, can drive the neurodegenerative process. The connection between different genes and proteinopathies through specific mechanisms has shed light on the pathophysiology of the disease, leading to the identification of potential pharmacological targets. New experimental strategies are emerging, in both preclinical and clinical settings, which focus on small molecules rather than gene therapy. In this review, we provide an insight into the aberrant mechanisms leading to FTLD-related proteinopathies and discuss recent therapies with the potential to ameliorate neurodegeneration and disease progression.
Collapse
Affiliation(s)
- Marcello Giunta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
37
|
Vissers MFJM, Heuberger JAAC, Groeneveld GJ. Targeting for Success: Demonstrating Proof-of-Concept with Mechanistic Early Phase Clinical Pharmacology Studies for Disease-Modification in Neurodegenerative Disorders. Int J Mol Sci 2021; 22:1615. [PMID: 33562713 PMCID: PMC7915613 DOI: 10.3390/ijms22041615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
The clinical failure rate for disease-modifying treatments (DMTs) that slow or stop disease progression has been nearly 100% for the major neurodegenerative disorders (NDDs), with many compounds failing in expensive and time-consuming phase 2 and 3 trials for lack of efficacy. Here, we critically review the use of pharmacological and mechanistic biomarkers in early phase clinical trials of DMTs in NDDs, and propose a roadmap for providing early proof-of-concept to increase R&D productivity in this field of high unmet medical need. A literature search was performed on published early phase clinical trials aimed at the evaluation of NDD DMT compounds using MESH terms in PubMed. Publications were selected that reported an early phase clinical trial with NDD DMT compounds between 2010 and November 2020. Attention was given to the reported use of pharmacodynamic (mechanistic and physiological response) biomarkers. A total of 121 early phase clinical trials were identified, of which 89 trials (74%) incorporated one or multiple pharmacodynamic biomarkers. However, only 65 trials (54%) used mechanistic (target occupancy or activation) biomarkers to demonstrate target engagement in humans. The most important categories of early phase mechanistic and response biomarkers are discussed and a roadmap for incorporation of a robust biomarker strategy for early phase NDD DMT clinical trials is proposed. As our understanding of NDDs is improving, there is a rise in potentially disease-modifying treatments being brought to the clinic. Further increasing the rational use of mechanistic biomarkers in early phase trials for these (targeted) therapies can increase R&D productivity with a quick win/fast fail approach in an area that has seen a nearly 100% failure rate to date.
Collapse
Affiliation(s)
- Maurits F. J. M. Vissers
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands; (J.A.A.C.H.); (G.J.G.)
- Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Jules A. A. C. Heuberger
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands; (J.A.A.C.H.); (G.J.G.)
| | - Geert Jan Groeneveld
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands; (J.A.A.C.H.); (G.J.G.)
- Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
38
|
Soeda Y, Takashima A. New Insights Into Drug Discovery Targeting Tau Protein. Front Mol Neurosci 2020; 13:590896. [PMID: 33343298 PMCID: PMC7744460 DOI: 10.3389/fnmol.2020.590896] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Microtubule-associated protein tau is characterized by the fact that it is an intrinsically disordered protein due to its lack of a stable conformation and high flexibility. Intracellular inclusions of fibrillar forms of tau with a β-sheet structure accumulate in the brain of patients with Alzheimer's disease and other tauopathies. Accordingly, detachment of tau from microtubules and transition of tau from a disordered state to an abnormally aggregated state are essential events preceding the onset of tau-related diseases. Many reports have shown that this transition is caused by post-translational modifications, including hyperphosphorylation and acetylation. The misfolded tau is self-assembled and forms a tau oligomer before the appearance of tau inclusions. Animal and pathological studies using human samples have demonstrated that tau oligomer formation contributes to neuronal loss. During the progression of tauopathies, tau seeds are released from cells and incorporated into other cells, leading to the propagation of pathological tau aggregation. Accumulating evidence suggests several potential approaches for blocking tau-mediated toxicity: (1) direct inhibition of pathological tau aggregation and (2) inhibition of tau post-translational modifications that occur prior to pathological tau aggregation, (3) inhibition of tau propagation and (4) stabilization of microtubules. In addition to traditional low-molecular-weight compounds, newer drug discovery approaches such as the development of medium-molecular-weight drugs (peptide- or oligonucleotide-based drugs) and high-molecular-weight drugs (antibody-based drugs) provide alternative pathways to preventing the formation of abnormal tau. Of particular interest are recent studies suggesting that tau droplet formation by liquid-liquid phase separation may be the initial step in aberrant tau aggregation, as well results that implicate roles for tau in dendritic and nuclear functions. Here, we review the mechanisms through which drugs can target tau and consider recent clinical trials for the treatment of tauopathies. In addition, we discuss the utility of these newer strategies and propose future directions for research on tau-targeted therapeutics.
Collapse
Affiliation(s)
- Yoshiyuki Soeda
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Akihiko Takashima
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| |
Collapse
|
39
|
Harris CJ, Gray NE, Caruso M, Hunter M, Ralle M, Quinn JF. Copper Modulation and Memory Impairment due to Hippocampal Tau Pathology. J Alzheimers Dis 2020; 78:49-60. [PMID: 32250309 DOI: 10.3233/jad-200002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background:Environmental copper has been implicated in the pathogenesis of Alzheimer’s disease based on evidence that: 1) brain copper levels increase with age, 2) copper promotes misfolding and toxicity of amyloid-β in vitro, 3) copper-modulating interventions reduce amyloid pathology in animal models. However, the effect of copper upon non-amyloid Alzheimer’s pathology is relatively under-explored.Objective:To determine if modulation of brain copper level affects brain tau pathology and/or associated cognitive impairment.Methods:We tested the hypothesis that brain copper modulates tau pathology by manipulating brain levels of copper in the PS19 transgenic mouse model of tau pathology. We treated PS19 and wild-type mice with oral zinc acetate, an established therapy for long term control of excess brain copper, and examined treatment effects upon brain copper, brain tau, NFT-like pathology, and spatial memory. We treated a second cohort of mice with exogenous dietary copper in order to evaluate whether excess environmental copper promotes brain tau pathology.Results:Copper-lowering with oral zinc attenuated spatial memory impairment in female but not male PS19 mice, without a significant effect upon tau pathology. Copper loading increased brain copper, but did not have an effect on brain tau pathology or spatial memory function.Conclusion:These findings suggest that a strategy to lower brain copper may be viable for symptomatic benefit in the setting of tau neuropathology, but unlikely to have robust effects on the underlying pathology. These findings are consistent with dietary or other exogenous copper being unlikely to promote tau pathology.
Collapse
Affiliation(s)
- Christopher J Harris
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Marguex Hunter
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Martina Ralle
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA.,Parkinsons Disease Research, Education, and Clinical Center, Portland Veterans Affairs Medical Center, Portland, OR, USA
| |
Collapse
|
40
|
VandeVrede L, Ljubenkov PA, Rojas JC, Welch AE, Boxer AL. Four-Repeat Tauopathies: Current Management and Future Treatments. Neurotherapeutics 2020; 17:1563-1581. [PMID: 32676851 PMCID: PMC7851277 DOI: 10.1007/s13311-020-00888-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Four-repeat tauopathies are a neurodegenerative disease characterized by brain parenchymal accumulation of a specific isoform of the protein tau, which gives rise to a wide breadth of clinical syndromes encompassing diverse symptomatology, with the most common syndromes being progressive supranuclear palsy-Richardson's and corticobasal syndrome. Despite the lack of effective disease-modifying therapies, targeted treatment of symptoms can improve quality of life for patients with 4-repeat tauopathies. However, managing these symptoms can be a daunting task, even for those familiar with the diseases, as they span motor, sensory, cognitive, affective, autonomic, and behavioral domains. This review describes current approaches to symptomatic management of common clinical symptoms in 4-repeat tauopathies with a focus on practical patient management, including pharmacologic and nonpharmacologic strategies, and concludes with a discussion of the history and future of disease-modifying therapeutics and clinical trials in this population.
Collapse
Affiliation(s)
- Lawren VandeVrede
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA.
| | - Peter A Ljubenkov
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ariane E Welch
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
41
|
VandeVrede L, Boxer AL, Polydoro M. Targeting tau: Clinical trials and novel therapeutic approaches. Neurosci Lett 2020; 731:134919. [PMID: 32380145 PMCID: PMC9212860 DOI: 10.1016/j.neulet.2020.134919] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 02/25/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022]
Abstract
Tauopathies are a group of over 20 clinicopathological neurodegenerative diseases including Alzheimer's disease (AD), the most common type of dementia, progressive supranuclear palsy, Pick's disease, corticobasal degeneration, among others. Tauopathies are defined by neurodegeneration and the presence of tau aggregates in affected brains regions. Interestingly, regional tau aggregation burden correlates with clinical phenotype and predicts cognitive status. Autosomal dominant mutations in the MAPT gene lead to tau deposition and clinical FTD syndromes with cognitive, behavioral, and motor impairment. Polymorphisms in or around the MAPT gene have also been strongly linked to other proteinopathies including synucleinopathies. Taken together these findings suggests that tau plays a critical role in neurodegeneration and proteinopathies, supporting the idea that tau targeted approaches can be disease-modifying and lead to clinically meaningful benefits in slowing or reversing disease progression. Increasingly, human clinical trials are testing this hypothesis. This article reviews tau-targeted therapies tested in clinical trials as well as agents currently in active development based on publicly disclosed information. We describe the therapeutic approaches of these trials based on the potential pathogenic mechanism they target.
Collapse
Affiliation(s)
- Lawren VandeVrede
- Memory and Aging Center, Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | | |
Collapse
|