1
|
Hagar HT, Fernandez-Vega V, Wang KW, Jordan LMO, Shumate J, Scampavia L, Tapayan AS, Nguyen HM, Spicer TP, Kuo MH. Hyperphosphorylated tau-based Alzheimer's Disease drug discovery: Identification of inhibitors of tau aggregation and cytotoxicity. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 33:100235. [PMID: 40319815 DOI: 10.1016/j.slasd.2025.100235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/14/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that affects more than 30 million people worldwide. Underlying the progressive decline of cognitive functions are the neurofibrillary tangles (NFTs) in neurons of the brain. The spatiotemporal distribution of NFTs predicts the progression of cognitive symptoms. In contrast, the senile plaques of amyloid-β aggregates, another major biomarker for AD, do not correlate with the clinical symptom development, consistent with the negligible benefits to cognitive functions in patients receiving anti-Aβ immunotherapies. A new drug discovery avenue targeting tau pathologies is therefore urgently needed. Using a recombinant hyperphosphorylated tau (p-tau) that presents characters key to the disease, e.g., formation of neurotoxic aggregates, we conducted a fluorescence p-tau aggregation assay and completed a 100K-compound high-throughput screen (HTS) and identified inhibitors of p-tau aggregation and cytotoxicity. This dual functional screen resulted in several potent compounds that effectively curbed both p-tau aggregation and cytotoxicity. Results presented in this work are the first HTS for small-molecule compounds that target the cellular toxicity of hyperphosphorylated tau. Top hits found in this screen and their analogues to be developed in the near future may lead to breakthroughs in the therapeutic development for Alzheimer's disease and other neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Hsiao-Tien Hagar
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Virneliz Fernandez-Vega
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology in Center, High-Throughput Molecular Screening Center, Department of Molecular Medicine, Jupiter, Florida 33458, USA
| | - Kuang-Wei Wang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Luis M Ortiz Jordan
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology in Center, High-Throughput Molecular Screening Center, Department of Molecular Medicine, Jupiter, Florida 33458, USA
| | - Justin Shumate
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology in Center, High-Throughput Molecular Screening Center, Department of Molecular Medicine, Jupiter, Florida 33458, USA
| | - Louis Scampavia
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology in Center, High-Throughput Molecular Screening Center, Department of Molecular Medicine, Jupiter, Florida 33458, USA
| | - April Sweet Tapayan
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, USA
| | - Timothy P Spicer
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology in Center, High-Throughput Molecular Screening Center, Department of Molecular Medicine, Jupiter, Florida 33458, USA.
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA.
| |
Collapse
|
2
|
Monaco M, Torazza C, Fedele E, Grilli M. The Impact of the Exposome on Alzheimer's Disease: The Influence of Nutrition. Int J Mol Sci 2025; 26:3015. [PMID: 40243652 PMCID: PMC11988514 DOI: 10.3390/ijms26073015] [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: 02/14/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline, memory loss, and behavioural changes. While genetic predispositions and pathological processes have been the traditional focus, this review highlights the fundamental role of environmental factors, particularly nutrition, within the exposome framework in modulating the risk and progression of AD. The exposome, which includes the totality of environmental exposures in an individual's lifetime, provides a comprehensive approach to understanding the complex aetiology of AD. In this review, we explore the impact of dietary factors and cyclic nucleotide pathways (cAMP/cGMP) on AD, emphasizing the potential of dietary interventions as therapeutic strategies. We investigate key aspects of how nutrition affects the accumulation of β-amyloid, the aggregation of tau proteins, and neuroinflammation. We also examine the impact of specific nutrients on cognitive performance and the risk of AD. Additionally, we discuss the potential of nutraceuticals with anti-phosphodiesterase activity and the role of various animal models of AD (such as 5xFAD, 3xTg-AD, Tg2576, and APP/PS1 mice) in demonstrating the effects of dietary interventions on disease onset and progression.
Collapse
Affiliation(s)
- Martina Monaco
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
| | - Carola Torazza
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
| | - Ernesto Fedele
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Massimo Grilli
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
3
|
Dickson JR, Sobolewski RGR, Fernandes AR, Cooper JM, Fan Z, Chung M, Donahue C, Oakley DH, Strickland DK, Hyman BT. Alzheimer disease-associated tau post-translational modification mimics impact tau propagation and uptake. J Neuropathol Exp Neurol 2025:nlaf007. [PMID: 39984820 DOI: 10.1093/jnen/nlaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025] Open
Abstract
As Alzheimer disease (AD) progresses, pathological tau spreads by cell-to-cell propagation of tau. This study aims to elucidate the impact of AD-associated post-translational modifications of tau-on-tau propagation. Tau propagation reporter constructs distinguishing donor cells from recipient cells were developed, and additional constructs were made with tau residues mutated from serine or threonine to aspartate to mimic the negative charge of a phosphorylation and/or from lysine to glutamine to mimic the charge-neutralizing effect of acetylation. Flow cytometry was used to quantify donor and recipient cells. This revealed that the mutations generally tended to reduce tau propagation compared to wildtype tau. Recombinant tau containing either wildtype or posttranslational modification mimicking mutations were used to treat Chinese hamster ovary cells or human induced pluripotent stem cell-derived neurons to quantify tau uptake, revealing that the mutations generally resulted in reduced uptake compared to wildtype tau. Surface plasmon resonance revealed that the mutations had a reduced affinity for lipoprotein receptor-related protein 1 (LRP1), a tau uptake receptor, compared to wildtype tau. Overall, these results suggest that AD-associated posttranslational modification mimicking mutations reduce the cell-to-cell propagation of tau by reducing tau uptake by recipient cells, which may be in part due to reduced binding affinity to LRP1.
Collapse
Affiliation(s)
- John R Dickson
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
- Faculty of Medicine, Harvard Medical School, Boston, MA, United States
| | - Robert G R Sobolewski
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Analiese R Fernandes
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Joanna M Cooper
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Zhanyun Fan
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Mirra Chung
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Cameron Donahue
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Derek H Oakley
- Faculty of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Pathology, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, United States
| | - Dudley K Strickland
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bradley T Hyman
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
- Faculty of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Feldman HH, Cummings JL, Boxer AL, Staffaroni AM, Knopman DS, Sukoff Rizzo SJ, Territo PR, Arnold SE, Ballard C, Beher D, Boeve BF, Dacks PA, Diaz K, Ewen C, Fiske B, Gonzalez MI, Harris GA, Hoffman BJ, Martinez TN, McDade E, Nisenbaum LK, Palma J, Quintana M, Rabinovici GD, Rohrer JD, Rosen HJ, Troyer MD, Kim DY, Tanzi RE, Zetterberg H, Ziogas NK, May PC, Rommel A. A framework for translating tauopathy therapeutics: Drug discovery to clinical trials. Alzheimers Dement 2024; 20:8129-8152. [PMID: 39316411 PMCID: PMC11567863 DOI: 10.1002/alz.14250] [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: 05/07/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/25/2024]
Abstract
The tauopathies are defined by pathological tau protein aggregates within a spectrum of clinically heterogeneous neurodegenerative diseases. The primary tauopathies meet the definition of rare diseases in the United States. There is no approved treatment for primary tauopathies. In this context, designing the most efficient development programs to translate promising targets and treatments from preclinical studies to early-phase clinical trials is vital. In September 2022, the Rainwater Charitable Foundation convened an international expert workshop focused on the translation of tauopathy therapeutics through early-phase trials. Our report on the workshop recommends a framework for principled drug development and a companion lexicon to facilitate communication focusing on reproducibility and achieving common elements. Topics include the selection of targets, drugs, biomarkers, participants, and study designs. The maturation of pharmacodynamic biomarkers to demonstrate target engagement and surrogate disease biomarkers is a crucial unmet need. HIGHLIGHTS: Experts provided a framework to translate therapeutics (discovery to clinical trials). Experts focused on the "5 Rights" (target, drug, biomarker, participants, trial). Current research on frontotemporal degeneration, progressive supranuclear palsy, and corticobasal syndrome therapeutics includes 32 trials (37% on biologics) Tau therapeutics are being tested in Alzheimer's disease; primary tauopathies have a large unmet need.
Collapse
Affiliation(s)
- Howard H. Feldman
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Jeffrey L. Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada at Las VegasLas VegasNevadaUSA
| | - Adam L. Boxer
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Adam M. Staffaroni
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | | | - Paul R. Territo
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Steven E. Arnold
- Department of NeurologyHarvard Medical SchoolMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Clive Ballard
- College of Medicine and HealthUniversity of ExeterExeterUK
| | | | | | - Penny A. Dacks
- The Association for Frontotemporal DegenerationKing of PrussiaPennsylvaniaUSA
| | | | | | - Brian Fiske
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | | | | | | | | | - Eric McDade
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Jose‐Alberto Palma
- Novartis Institutes for Biomedical ResearchCambridgeMassachusettsUSA
- Department of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | | | - Gil D. Rabinovici
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jonathan D. Rohrer
- Department of Neurodegenerative DiseaseDementia Research CentreQueen Square Institute of NeurologyUniversity College of LondonLondonUK
| | - Howard J. Rosen
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Doo Yeon Kim
- Department of NeurologyGenetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Rudolph E. Tanzi
- Department of NeurologyGenetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistrySahlgrenska Academy at the University of GothenburgMölndalSweden
| | | | - Patrick C. May
- ADvantage Neuroscience Consulting LLCFort WayneIndianaUSA
| | - Amy Rommel
- Rainwater Charitable FoundationFort WorthTexasUSA
| |
Collapse
|
5
|
Ayers J, Lopez TP, Steele IT, Oehler A, Roman-Albarran R, Cleveland E, Chong A, Carlson GA, Condello C, Prusiner SB. Severe neurodegeneration in brains of transgenic rats producing human tau prions. Acta Neuropathol 2024; 148:25. [PMID: 39160375 PMCID: PMC11333523 DOI: 10.1007/s00401-024-02771-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024]
Abstract
Both wild-type and mutant tau proteins can misfold into prions and self-propagate in the central nervous system of animals and people. To extend the work of others, we investigated the molecular basis of tau prion-mediated neurodegeneration in transgenic (Tg) rats expressing mutant human tau (P301S); this line of Tg rats is denoted Tg12099. We used the rat Prnp promoter to drive the overexpression of mutant tau (P301S) in the human 0N4R isoform. In Tg12099(+/+) rats homozygous for the transgene, ubiquitous expression of mutant human tau resulted in the progressive accumulation of phosphorylated tau inclusions, including silver-positive tangles in the frontal cortices and limbic system. Signs of central nervous system dysfunction were found in terminal Tg12099(+/+) rats exhibiting severe neurodegeneration and profound atrophy of the amygdala and piriform cortex. The greatest increases in tau prion activity were found in the corticolimbic structures. In contrast to the homozygous Tg12099(+/+) rats, we found lower levels of mutant tau in the hemizygous rats, resulting in few neuropathologic changes up to 2 years of age. Notably, these hemizygous rats could be infected by intracerebral inoculation with recombinant tau fibrils or precipitated tau prions from the brain homogenates of sick, aged homozygous Tg12099(+/+) rats. Our studies argue that the regional propagation of tau prions and neurodegeneration in the Tg12099 rats resembles that found in human primary tauopathies. These findings seem likely to advance our understanding of human tauopathies and may lead to effective therapeutics for Alzheimer's disease and other tau prion disorders.
Collapse
Affiliation(s)
- Jacob Ayers
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - T Peter Lopez
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Ian T Steele
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Abby Oehler
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Rigo Roman-Albarran
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Elisa Cleveland
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Alex Chong
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - George A Carlson
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Carlo Condello
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
6
|
Nishida I, Yamada K, Sakamoto A, Wakabayashi T, Iwatsubo T. Chronic Neuronal Hyperexcitation Exacerbates Tau Propagation in a Mouse Model of Tauopathy. Int J Mol Sci 2024; 25:9004. [PMID: 39201689 PMCID: PMC11354494 DOI: 10.3390/ijms25169004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
The intracerebral spread of tau is a critical mechanism associated with functional decline in Alzheimer's disease (AD) and other tauopathies. Recently, a hypothesis has emerged suggesting that tau propagation is linked to functional neuronal connections, specifically driven by neuronal hyperactivity. However, experimental validation of this hypothesis remains limited. In this study, we investigated how tau propagation from the entorhinal cortex to the hippocampus, the neuronal circuit most susceptible to tau pathology in AD, is affected by the selective stimulation of neuronal activity along this circuit. Using a mouse model of seed-induced propagation combined with optogenetics, we found that the chronic stimulation of this neuronal connection over a 4-week period resulted in a significant increase in insoluble tau accumulation in both the entorhinal cortex and hippocampus. Importantly, the ratio of tau accumulation in the hippocampus relative to that in the entorhinal cortex, serving as an indicator of transcellular spreading, was significantly higher in mice subjected to chronic stimulation. These results support the notion that abnormal neuronal activity promotes tau propagation, thereby implicating it in the progression of tauopathy.
Collapse
Affiliation(s)
- Itaru Nishida
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Asami Sakamoto
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Tomoko Wakabayashi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
- Department of Pathophysiology, Meiji Pharmaceutical University, Tokyo 2040004, Japan
| | - Takeshi Iwatsubo
- National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 1878551, Japan;
| |
Collapse
|
7
|
Catterson JH, Mouofo EN, López De Toledo Soler I, Lean G, Dlamini S, Liddell P, Voong G, Katsinelos T, Wang YC, Schoovaerts N, Verstreken P, Spires-Jones TL, Durrant CS. Drosophila appear resistant to trans-synaptic tau propagation. Brain Commun 2024; 6:fcae256. [PMID: 39130515 PMCID: PMC11316205 DOI: 10.1093/braincomms/fcae256] [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: 03/11/2024] [Revised: 05/22/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024] Open
Abstract
Alzheimer's disease is the most common cause of dementia in the elderly, prompting extensive efforts to pinpoint novel therapeutic targets for effective intervention. Among the hallmark features of Alzheimer's disease is the development of neurofibrillary tangles comprised of hyperphosphorylated tau protein, whose progressive spread throughout the brain is associated with neuronal death. Trans-synaptic propagation of tau has been observed in mouse models, and indirect evidence for tau spread via synapses has been observed in human Alzheimer's disease. Halting tau propagation is a promising therapeutic target for Alzheimer's disease; thus, a scalable model system to screen for modifiers of tau spread would be very useful for the field. To this end, we sought to emulate the trans-synaptic spread of human tau in Drosophila melanogaster. Employing the trans-Tango circuit mapping technique, we investigated whether tau spreads between synaptically connected neurons. Immunohistochemistry and confocal imaging were used to look for tau propagation. Examination of hundreds of flies expressing four different human tau constructs in two distinct neuronal populations reveals a robust resistance in Drosophila to the trans-synaptic spread of human tau. This resistance persisted in lines with concurrent expression of amyloid-β, in lines with global human tau knock-in to provide a template for human tau in downstream neurons, and with manipulations of temperature. These negative data are important for the field as we establish that Drosophila expressing human tau in subsets of neurons are unlikely to be useful to perform screens to find mechanisms to reduce the trans-synaptic spread of tau. The inherent resistance observed in Drosophila may serve as a valuable clue, offering insights into strategies for impeding tau spread in future studies.
Collapse
Affiliation(s)
- James H Catterson
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Edmond N Mouofo
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | | | - Gillian Lean
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Stella Dlamini
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Phoebe Liddell
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Graham Voong
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Taxiarchis Katsinelos
- Schaller Research Group at the University of Heidelberg and the DKFZ, German Cancer Research Center, Proteostasis in Neurodegenerative Disease (B180), INF 581, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, INF 234, 69120 Heidelberg, Germany
| | - Yu-Chun Wang
- VIB-KU Leuven Center for Brain & Disease Research, Department of Neurosciences, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Nils Schoovaerts
- VIB-KU Leuven Center for Brain & Disease Research, Department of Neurosciences, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, Department of Neurosciences, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Claire S Durrant
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh EH8 9XD, UK
| |
Collapse
|
8
|
Gorini F, Tonacci A. Metal Toxicity and Dementia Including Frontotemporal Dementia: Current State of Knowledge. Antioxidants (Basel) 2024; 13:938. [PMID: 39199184 PMCID: PMC11351151 DOI: 10.3390/antiox13080938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
Frontotemporal dementia (FTD) includes a number of neurodegenerative diseases, often with early onset (before 65 years old), characterized by progressive, irreversible deficits in behavioral, linguistic, and executive functions, which are often difficult to diagnose due to their similar phenotypic characteristics to other dementias and psychiatric disorders. The genetic contribution is of utmost importance, although environmental risk factors also play a role in its pathophysiology. In fact, some metals are known to produce free radicals, which, accumulating in the brain over time, can induce oxidative stress, inflammation, and protein misfolding, all of these being key features of FTD and similar conditions. Therefore, the present review aims to summarize the current evidence about the environmental contribution to FTD-mainly dealing with toxic metal exposure-since the identification of such potential environmental risk factors can lead to its early diagnosis and the promotion of policies and interventions. This would allow us, by reducing exposure to these pollutants, to potentially affect society at large in a positive manner, decreasing the burden of FTD and similar conditions on affected individuals and society overall. Future perspectives, including the application of Artificial Intelligence principles to the field, with related evidence found so far, are also introduced.
Collapse
Affiliation(s)
| | - Alessandro Tonacci
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| |
Collapse
|
9
|
Zubčić K, Franić D, Pravica M, Hof PR, Šimić G, Boban M. Effects of heterologous human tau protein expression in yeast models of proteotoxic stress response. CNS Neurosci Ther 2024; 30:e14304. [PMID: 37341072 PMCID: PMC11163194 DOI: 10.1111/cns.14304] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND The primary histological characteristic of Alzheimer's disease is the presence of neurofibrillary tangles, which are large aggregates of tau protein. Aging is the primary risk factor for the development of Alzheimer's disease, however, the underlying causes of tau protein aggregation and toxicity are unclear. AIMS Here we investigated tau aggregation and toxicity under the conditions of compromised protein homeostasis. METHODS We used heterologous expression of human tau protein in the unicellular eukaryote yeast Saccharomyces cerevisiae with evolutionarily conserved protein quality control pathways and examined tau-dependent toxicity and aggregation using growth assays, fluorescence microscopy, and a split luciferase-based reporter NanoBiT. RESULTS Tau protein expressed in yeast under mild proteotoxic stress, or in mutants with impaired pathways for proteotoxic stress response, did not lead to synthetic toxicity or the formation of obvious aggregates. Chronologically old cells also did not develop observable tau aggregates. Our examination of tau oligomerization in living cells using NanoBiT reporter suggests that tau does not form significant levels of oligomers under basal conditions or under mild proteotoxic stress. CONCLUSION Together our data suggest that human tau protein does not represent a major burden to the protein quality control system in yeast cells.
Collapse
Affiliation(s)
- Klara Zubčić
- Croatian Institute for Brain Research, University of Zagreb School of MedicineZagrebCroatia
| | - Dina Franić
- Croatian Institute for Brain Research, University of Zagreb School of MedicineZagrebCroatia
| | - Mihaela Pravica
- Croatian Institute for Brain Research, University of Zagreb School of MedicineZagrebCroatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's DiseaseFriedman Brain Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Goran Šimić
- Croatian Institute for Brain Research, University of Zagreb School of MedicineZagrebCroatia
| | - Mirta Boban
- Croatian Institute for Brain Research, University of Zagreb School of MedicineZagrebCroatia
| |
Collapse
|
10
|
Boccardi V, Orr ME, Polidori MC, Ruggiero C, Mecocci P. Focus on senescence: Clinical significance and practical applications. J Intern Med 2024; 295:599-619. [PMID: 38446642 DOI: 10.1111/joim.13775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The older population is increasing worldwide, and life expectancy is continuously rising, predominantly thanks to medical and technological progress. Healthspan refers to the number of years an individual can live in good health. From a gerontological viewpoint, the mission is to extend the life spent in good health, promoting well-being and minimizing the impact of aging-related diseases to slow the aging process. Biologically, aging is a malleable process characterized by an intra- and inter-individual heterogeneous and dynamic balance between accumulating damage and repair mechanisms. Cellular senescence is a key component of this process, with senescent cells accumulating in different tissues and organs, leading to aging and age-related disease susceptibility over time. Removing senescent cells from the body or slowing down the burden rate has been proposed as an efficient way to reduce age-dependent deterioration. In animal models, senotherapeutic molecules can extend life expectancy and lifespan by either senolytic or senomorphic activity. Much research shows that dietary and physical activity-driven lifestyle interventions protect against senescence. This narrative review aims to summarize the current knowledge on targeting senescent cells to reduce the risk of age-related disease in animal models and their translational potential for humans. We focused on studies that have examined the potential role of senotherapeutics in slowing the aging process and modifying age-related disease burdens. The review concludes with a general discussion of the mechanisms underlying this unique trajectory and its implications for future research.
Collapse
Affiliation(s)
- Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Miranda Ethel Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Salisbury VA Medical Center, Salisbury, North Carolina, USA
| | - M Cristina Polidori
- Ageing Clinical Research, Department II of Internal Medicine and Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress-Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carmelinda Ruggiero
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Patrizia Mecocci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Granholm AC, Hamlett ED. The Role of Tau Pathology in Alzheimer's Disease and Down Syndrome. J Clin Med 2024; 13:1338. [PMID: 38592182 PMCID: PMC10932364 DOI: 10.3390/jcm13051338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Individuals with Down syndrome (DS) exhibit an almost complete penetrance of Alzheimer's disease (AD) pathology but are underrepresented in clinical trials for AD. The Tau protein is associated with microtubule function in the neuron and is crucial for normal axonal transport. In several different neurodegenerative disorders, Tau misfolding leads to hyper-phosphorylation of Tau (p-Tau), which may seed pathology to bystander cells and spread. This review is focused on current findings regarding p-Tau and its potential to seed pathology as a "prion-like" spreader. It also considers the consequences of p-Tau pathology leading to AD, particularly in individuals with Down syndrome. Methods: Scopus (SC) and PubMed (PM) were searched in English using keywords "tau AND seeding AND brain AND down syndrome". A total of 558 SC or 529 PM potentially relevant articles were identified, of which only six SC or three PM articles mentioned Down syndrome. This review was built upon the literature and the recent findings of our group and others. Results: Misfolded p-Tau isoforms are seeding competent and may be responsible for spreading AD pathology. Conclusions: This review demonstrates recent work focused on understanding the role of neurofibrillary tangles and monomeric/oligomeric Tau in the prion-like spreading of Tau pathology in the human brain.
Collapse
Affiliation(s)
- Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Eric D. Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
12
|
Rogers BB, Anderson AG, Lauzon SN, Davis MN, Hauser RM, Roberts SC, Rodriguez-Nunez I, Trausch-Lowther K, Barinaga EA, Hall PI, Knuesel MT, Taylor JW, Mackiewicz M, Roberts BS, Cooper SJ, Rizzardi LF, Myers RM, Cochran JN. Neuronal MAPT expression is mediated by long-range interactions with cis-regulatory elements. Am J Hum Genet 2024; 111:259-279. [PMID: 38232730 PMCID: PMC10870142 DOI: 10.1016/j.ajhg.2023.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
Tauopathies are a group of neurodegenerative diseases defined by abnormal aggregates of tau, a microtubule-associated protein encoded by MAPT. MAPT expression is near absent in neural progenitor cells (NPCs) and increases during differentiation. This temporally dynamic expression pattern suggests that MAPT expression could be controlled by transcription factors and cis-regulatory elements specific to differentiated cell types. Given the relevance of MAPT expression to neurodegeneration pathogenesis, identification of such elements is relevant to understanding disease risk and pathogenesis. Here, we performed chromatin conformation assays (HiC & Capture-C), single-nucleus multiomics (RNA-seq+ATAC-seq), bulk ATAC-seq, and ChIP-seq for H3K27ac and CTCF in NPCs and differentiated neurons to nominate candidate cis-regulatory elements (cCREs). We assayed these cCREs using luciferase assays and CRISPR interference (CRISPRi) experiments to measure their effects on MAPT expression. Finally, we integrated cCRE annotations into an analysis of genetic variation in neurodegeneration-affected individuals and control subjects. We identified both proximal and distal regulatory elements for MAPT and confirmed the regulatory function for several regions, including three regions centromeric to MAPT beyond the H1/H2 haplotype inversion breakpoint. We also found that rare and predicted damaging genetic variation in nominated CREs was nominally depleted in dementia-affected individuals relative to control subjects, consistent with the hypothesis that variants that disrupt MAPT enhancer activity, and thereby reduced MAPT expression, may be protective against neurodegenerative disease. Overall, this study provides compelling evidence for pursuing detailed knowledge of CREs for genes of interest to permit better understanding of disease risk.
Collapse
Affiliation(s)
- Brianne B Rogers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Shelby N Lauzon
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - M Natalie Davis
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Rebecca M Hauser
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Sydney C Roberts
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | | | - Erin A Barinaga
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Paige I Hall
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Jared W Taylor
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Mark Mackiewicz
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Brian S Roberts
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Sara J Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA.
| | | |
Collapse
|
13
|
Sun X, Ogbolu VC, Baas PW, Qiang L. Reevaluating tau reduction as a therapeutic approach for tauopathies: Insights and perspectives. Cytoskeleton (Hoboken) 2024; 81:57-62. [PMID: 37819557 PMCID: PMC10843461 DOI: 10.1002/cm.21790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023]
Abstract
Tau, one of the most abundant microtubule-associated protein in neurons plays a role in regulating microtubule dynamics in axons, as well as shaping the overall morphology of the axon. Recent studies challenge the traditional view of tau as a microtubule stabilizer and shed new light on the complexity of its role in regulating various properties of the microtubule. While reducing tau levels shows therapeutic promise for early tauopathies, efficacy wanes in later stages due to resilient toxic tau aggregates and neurofibrillary tangles. Notably, tauopathies involve factors beyond toxic tau alone, necessitating a broader therapeutic approach. Overexpression of human tau in mouse models, although useful for answering some questions, may not accurately reflect disease mechanisms in patients with tauopathies. Furthermore, the interplay between tau and MAP6, another microtubule-associated protein, adds complexity to tau's regulation of microtubule dynamics. Tau promotes the formation and elongation of labile microtubule domains, vital for cellular processes, while MAP6 stabilizes microtubules. A delicate balance between these proteins is important for neuronal function. Therefore, tau reduction therapies require a comprehensive understanding of disease progression, considering functional tau loss, toxic aggregates, and microtubule dynamics. Stage-dependent application and potential unintended consequences must be carefully evaluated. Restoring microtubule dynamics in late-stage tauopathies may necessitate alternative strategies. This knowledge is valuable for developing effective and safe treatments for tauopathies.
Collapse
Affiliation(s)
- Xiaohuan Sun
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Victor C. Ogbolu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Peter W. Baas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Liang Qiang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| |
Collapse
|
14
|
Hu J, Sha W, Yuan S, Wu J, Huang Y. Aggregation, Transmission, and Toxicity of the Microtubule-Associated Protein Tau: A Complex Comprehension. Int J Mol Sci 2023; 24:15023. [PMID: 37834471 PMCID: PMC10573976 DOI: 10.3390/ijms241915023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The microtubule-associated protein tau is an intrinsically disordered protein containing a few short and transient secondary structures. Tau physiologically associates with microtubules (MTs) for its stabilization and detaches from MTs to regulate its dynamics. Under pathological conditions, tau is abnormally modified, detaches from MTs, and forms protein aggregates in neuronal and glial cells. Tau protein aggregates can be found in a number of devastating neurodegenerative diseases known as "tauopathies", such as Alzheimer's disease (AD), frontotemporal dementia (FTD), corticobasal degeneration (CBD), etc. However, it is still unclear how the tau protein is compacted into ordered protein aggregates, and the toxicity of the aggregates is still debated. Fortunately, there has been considerable progress in the study of tau in recent years, particularly in the understanding of the intercellular transmission of pathological tau species, the structure of tau aggregates, and the conformational change events in the tau polymerization process. In this review, we summarize the concepts of tau protein aggregation and discuss the views on tau protein transmission and toxicity.
Collapse
Affiliation(s)
- Jiaxin Hu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
| | - Wenchi Sha
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
| | - Shuangshuang Yuan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
| | - Jiarui Wu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yunpeng Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; (J.H.); (W.S.); (S.Y.)
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
15
|
Rodrigues S, Anglada-Huguet M, Hochgräfe K, Kaniyappan S, Wegmann S, Mandelkow EM. Spreading of Tau Protein Does Not Depend on Aggregation Propensity. J Mol Neurosci 2023; 73:693-712. [PMID: 37606769 PMCID: PMC10694122 DOI: 10.1007/s12031-023-02143-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/10/2023] [Indexed: 08/23/2023]
Abstract
The stereotypical progression of Tau pathology during Alzheimer disease has been attributed to trans-neuronal spreading of misfolded Tau proteins, followed by prion-like templated aggregation of Tau. The nature of Tau and the cellular mechanisms of Tau spreading are still under debate. We hypothesized that Tau's propensity for aggregation would correlate with its ability to spread across synapses and propagate pathology. To study the progressive propagation of Tau proteins in brain regions relevant for Alzheimer disease, we used mice expressing near-physiological levels of full-length human Tau protein carrying pro-aggregant (TauΔK280, TauΔK) or anti-aggregant (TauΔK280-PP, TauΔK-PP) mutations in the entorhinal cortex (EC). To enhance Tau expression in the EC, we performed EC injections of adeno-associated virus (AAV) particles encoding TauΔK or TauΔK-PP. The brains of injected and non-injected EC/TauΔK and EC/TauΔK-PP mice were studied by immunohistological and biochemical techniques to detect Tau propagation to dentate gyrus (DG) neurons and Tau-induced pathological changes. Pro- and anti-aggregant mice had comparable low transgene expression (~0.2 times endogenous mouse Tau). They accumulated human Tau at similar rates and only in expressing EC neurons, including their axonal projections of the perforant path and presynaptic terminals in the molecular layer of the DG. Pro-aggregant EC/TauΔK mice showed misfolded Tau and synaptic protein alterations in EC neurons, not observed in anti-aggregant EC/TauΔK-PP mice. Additional AAV-mediated expression of TauΔK or TauΔK-PP in EC/TauΔK or EC/TauΔK-PP mice, respectively, increased the human Tau expression to ~0.65 times endogenous mouse Tau, with comparable spreading of TauΔK and TauΔK-PP throughout the EC. There was a low level of transcellular propagation of Tau protein, without pathological phosphorylation or misfolding, as judged by diagnostic antibodies. Additionally, TauΔK but not TauΔK-PP expression induced hippocampal astrogliosis. Low levels of pro- or anti-aggregant full-length Tau show equivalent distributions in EC neurons, independent of their aggregation propensity. Increasing the expression via AAV induce local Tau misfolding in the EC neurons, synaptotoxicity, and astrogliosis and lead to a low level of detectable trans-neuronal spreading of Tau. This depends on its concentration in the EC, but, contrary to expectations, does not depend on Tau's aggregation propensity/misfolding and does not lead to templated misfolding in recipient neurons.
Collapse
Affiliation(s)
- Sara Rodrigues
- DZNE, German Ctr. for Neurodegenerative Diseases, Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Marta Anglada-Huguet
- DZNE, German Ctr. for Neurodegenerative Diseases, Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Katja Hochgräfe
- DZNE, German Ctr. for Neurodegenerative Diseases, Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Senthilvelrajan Kaniyappan
- DZNE, German Ctr. for Neurodegenerative Diseases, Venusberg-Campus 1/99, 53127, Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical School, Bonn, Germany
| | - Susanne Wegmann
- DZNE, German Center for Neurodegenerative Diseases, Chariteplatz 1, 10117, Berlin, Germany
| | - Eva-Maria Mandelkow
- DZNE, German Ctr. for Neurodegenerative Diseases, Venusberg-Campus 1/99, 53127, Bonn, Germany.
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
| |
Collapse
|
16
|
Morón-Oset J, Fischer LKS, Jauré N, Zhang P, Jahn AJ, Supèr T, Pahl A, Isaacs AM, Grönke S, Partridge L. Repeat length of C9orf72-associated glycine-alanine polypeptides affects their toxicity. Acta Neuropathol Commun 2023; 11:140. [PMID: 37644512 PMCID: PMC10463776 DOI: 10.1186/s40478-023-01634-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/31/2023] Open
Abstract
G4C2 hexanucleotide repeat expansions in a non-coding region of the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). G4C2 insertion length is variable, and patients can carry up to several thousand repeats. Dipeptide repeat proteins (DPRs) translated from G4C2 transcripts are thought to be a main driver of toxicity. Experiments in model organisms with relatively short DPRs have shown that arginine-rich DPRs are most toxic, while polyGlycine-Alanine (GA) DPRs cause only mild toxicity. However, GA is the most abundant DPR in patient brains, and experimental work in animals has generally relied on the use of low numbers of repeats, with DPRs often tagged for in vivo tracking. Whether repeat length or tagging affect the toxicity of GA has not been systematically assessed. Therefore, we generated Drosophila fly lines expressing GA100, GA200 or GA400 specifically in adult neurons. Consistent with previous studies, expression of GA100 and GA200 caused only mild toxicity. In contrast, neuronal expression of GA400 drastically reduced climbing ability and survival of flies, indicating that long GA DPRs can be highly toxic in vivo. This toxicity could be abolished by tagging GA400. Proteomics analysis of fly brains showed a repeat-length-dependent modulation of the brain proteome, with GA400 causing earlier and stronger changes than shorter GA proteins. PolyGA expression up-regulated proteins involved in ER to Golgi trafficking, and down-regulated proteins involved in insulin signalling. Experimental down-regulation of Tango1, a highly conserved regulator of ER-to Golgi transport, partially rescued GA400 toxicity, suggesting that misregulation of this process contributes to polyGA toxicity. Experimentally increasing insulin signaling also rescued GA toxicity. In summary, our data show that long polyGA proteins can be highly toxic in vivo, and that they may therefore contribute to ALS/FTD pathogenesis in patients.
Collapse
Affiliation(s)
- Javier Morón-Oset
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | | | - Nathalie Jauré
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Pingze Zhang
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Annika Julia Jahn
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Tessa Supèr
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - André Pahl
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Sebastian Grönke
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany.
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany.
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
17
|
Chen Y, Yu Y. Tau and neuroinflammation in Alzheimer's disease: interplay mechanisms and clinical translation. J Neuroinflammation 2023; 20:165. [PMID: 37452321 PMCID: PMC10349496 DOI: 10.1186/s12974-023-02853-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Alzheimer's Disease (AD) contributes to most cases of dementia. Its prominent neuropathological features are the extracellular neuritic plaques and intercellular neurofibrillary tangles composed of aggregated β-amyloid (Aβ) and hyperphosphorylated tau protein, respectively. In the past few decades, disease-modifying therapy targeting Aβ has been the focus of AD drug development. Even though it is encouraging that two of these drugs have recently received accelerated US Food and Drug Administration approval for AD treatment, their efficacy or long-term safety is controversial. Tau has received increasing attention as a potential therapeutic target, since evidence indicates that tau pathology is more associated with cognitive dysfunction. Moreover, inflammation, especially neuroinflammation, accompanies AD pathological processes and is also linked to cognitive deficits. Accumulating evidence indicates that inflammation has a complex and tight interplay with tau pathology. Here, we review recent evidence on the interaction between tau pathology, focusing on tau post-translational modification and dissemination, and neuroinflammatory responses, including glial cell activation and inflammatory signaling pathways. Then, we summarize the latest clinical trials targeting tau and neuroinflammation. Sustained and increased inflammatory responses in glial cells and neurons are pivotal cellular drivers and regulators of the exacerbation of tau pathology, which further contributes to its worsening by aggravating inflammatory responses. Unraveling the precise mechanisms underlying the relationship between tau pathology and neuroinflammation will provide new insights into the discovery and clinical translation of therapeutic targets for AD and other tau-related diseases (tauopathies). Targeting multiple pathologies and precision therapy strategies will be the crucial direction for developing drugs for AD and other tauopathies.
Collapse
Affiliation(s)
- Yijun Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
18
|
Rogers BB, Anderson AG, Lauzon SN, Davis MN, Hauser RM, Roberts SC, Rodriguez-Nunez I, Trausch-Lowther K, Barinaga EA, Taylor JW, Mackiewicz M, Roberts BS, Cooper SJ, Rizzardi LF, Myers RM, Cochran JN. MAPT expression is mediated by long-range interactions with cis-regulatory elements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531520. [PMID: 37090552 PMCID: PMC10120716 DOI: 10.1101/2023.03.07.531520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Background Tauopathies are a group of neurodegenerative diseases driven by abnormal aggregates of tau, a microtubule associated protein encoded by the MAPT gene. MAPT expression is absent in neural progenitor cells (NPCs) and increases during differentiation. This temporally dynamic expression pattern suggests that MAPT expression is controlled by transcription factors and cis-regulatory elements specific to differentiated cell types. Given the relevance of MAPT expression to neurodegeneration pathogenesis, identification of such elements is relevant to understanding genetic risk factors. Methods We performed HiC, chromatin conformation capture (Capture-C), single-nucleus multiomics (RNA-seq+ATAC-seq), bulk ATAC-seq, and ChIP-seq for H3K27Ac and CTCF in NPCs and neurons differentiated from human iPSC cultures. We nominated candidate cis-regulatory elements (cCREs) for MAPT in human NPCs, differentiated neurons, and pure cultures of inhibitory and excitatory neurons. We then assayed these cCREs using luciferase assays and CRISPR interference (CRISPRi) experiments to measure their effects on MAPT expression. Finally, we integrated cCRE annotations into an analysis of genetic variation in AD cases and controls. Results Using orthogonal genomics approaches, we nominated 94 cCREs for MAPT, including the identification of cCREs specifically active in differentiated neurons. Eleven regions enhanced reporter gene transcription in luciferase assays. Using CRISPRi, 5 of the 94 regions tested were identified as necessary for MAPT expression as measured by RT-qPCR and RNA-seq. Rare and predicted damaging genetic variation in both nominated and confirmed CREs was depleted in AD cases relative to controls (OR = 0.40, p = 0.004), consistent with the hypothesis that variants that disrupt MAPT enhancer activity, and thereby reduce MAPT expression, may be protective against neurodegenerative disease. Conclusions We identified both proximal and distal regulatory elements for MAPT and confirmed the regulatory function for several regions, including three regions centromeric to MAPT beyond the well-described H1/H2 haplotype inversion breakpoint. This study provides compelling evidence for pursuing detailed knowledge of CREs for genes of interest to permit better understanding of disease risk.
Collapse
Affiliation(s)
- Brianne B. Rogers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | | | | | | | - Jared W. Taylor
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Mark Mackiewicz
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - Sara J. Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | | | | |
Collapse
|
19
|
Moore KBE, Hung TJ, Fortin JS. Hyperphosphorylated tau (p-tau) and drug discovery in the context of Alzheimer's disease and related tauopathies. Drug Discov Today 2023; 28:103487. [PMID: 36634842 PMCID: PMC9975055 DOI: 10.1016/j.drudis.2023.103487] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by intracellular neurofibrillary tangles (NFTs) and extracellular β-amyloid (βA) plaques. No disease-modifying therapy is currently available to prevent the progression of, or cure, the disease. Misfolded hyperphosphorylated tau (p-tau) is considered a pivotal point in the pathogenesis of AD and other tauopathies. Compelling evidence suggests that it is a key driver of the accumulation of NFTs and can be directly correlated with the extent of dementia in patients with AD. Therefore, inhibiting tau hyperphosphorylation-induced aggregation could be a viable strategy to discover and develop therapeutics for patients with AD.
Collapse
Affiliation(s)
- Kendall B E Moore
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA
| | - Ta-Jung Hung
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA
| | - Jessica S Fortin
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA.
| |
Collapse
|
20
|
Kim SH, Lim KH, Yang S, Joo JY. Boosting of tau protein aggregation by CD40 and CD48 gene expression in Alzheimer's disease. FASEB J 2023; 37:e22702. [PMID: 36520044 DOI: 10.1096/fj.202201197r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/09/2022] [Accepted: 11/28/2022] [Indexed: 12/16/2022]
Abstract
Neurodegenerative diseases result from the interplay of abnormal gene expression and various pathological factors. Therefore, a disease-specific integrative genetic approach is required to understand the complexities and causes of target diseases. Recent studies have identified the correlation between genes encoding several transmembrane proteins, such as the cluster of differentiation (CD) and Alzheimer's disease (AD) pathogenesis. In this study, CD48 and CD40 gene expression in AD, a neurodegenerative disease, was analyzed to infer this link. Total RNA sequencing was performed using an Alzheimer's disease mouse model brain and blood, and gene expression was determined using a genome-wide association study (GWAS). We observed a marked elevation of CD48 and CD40 genes in Alzheimer's disease. Indeed, the upregulation of both CD48 and CD40 genes was significantly increased in the severe Alzheimer's disease group. With the elevation of CD48 and CD40 genes in Alzheimer's disease, associations of protein levels were also markedly increased in tissues. In addition, overexpression of CD48 and CD40 genes triggered tau aggregation, and co-expression of these genes accelerated aggregation. The nuclear factor kappa B (NF-ĸB) signaling pathway was enriched by CD48 and CD40 gene expression: it was also associated with tau pathology. Our data suggested that the CD48 and CD40 genes are novel AD-related genes, and this approach may be useful as a diagnostic or therapeutic target for the disease.
Collapse
Affiliation(s)
- Sung-Hyun Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Republic of Korea
| | - Key-Hwan Lim
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, Republic of Korea.,Department of Pharmacy, College of Pharmacy, Chungbuk National University, Cheongju-si, Republic of Korea
| | - Sumin Yang
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Republic of Korea
| | - Jae-Yeol Joo
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
21
|
Davies C, Tulloch J, Yip E, Currie L, Colom-Cadena M, Wegmann S, Hyman BT, Wilkins L, Hooley M, Tzioras M, Spires-Jones TL. Apolipoprotein E isoform does not influence trans-synaptic spread of tau pathology in a mouse model. Brain Neurosci Adv 2023; 7:23982128231191046. [PMID: 37600228 PMCID: PMC10433884 DOI: 10.1177/23982128231191046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/23/2023] [Indexed: 08/22/2023] Open
Abstract
A key hallmark of Alzheimer's disease (AD) is the accumulation of hyperphosphorylated tau in neurofibrillary tangles. This occurs alongside neuroinflammation and neurodegeneration. Pathological tau propagates through the AD brain in a defined manner, which correlates with neuron and synapse loss and cognitive decline. One proposed mechanism of tau spread is through synaptically connected brain structures. Apolipoprotein E4 (APOE4) genotype is the strongest genetic risk factor for late-onset AD and is associated with increased tau burden. Whether the apolipoprotein E (APOE) genotype influences neurodegeneration via tau spread is currently unknown. Here, we demonstrate that virally expressed human tau (with the P301L mutation) injected into mouse entorhinal cortex at 5-6 months or 15-16 months of age spreads trans-synaptically to the hippocampus by 14 weeks post-injection. Injections of tau in mice expressing human APOE2, APOE3 or APOE4, as well as APOE knock-outs, showed that tau can spread trans-synaptically in all genotypes and that APOE genotype and age do not affect the spread of tau. These data suggest that APOE genotype is not directly linked to synaptic spread of tau in our model, but other mechanisms involving non-cell autonomous manners of tau spread are still possible.
Collapse
Affiliation(s)
- Caitlin Davies
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jane Tulloch
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Ellie Yip
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Lydia Currie
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Marti Colom-Cadena
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lewis Wilkins
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Monique Hooley
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Makis Tzioras
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
22
|
The Strategies for Treating "Alzheimer's Disease": Insulin Signaling May Be a Feasible Target. Curr Issues Mol Biol 2022; 44:6172-6188. [PMID: 36547082 PMCID: PMC9777526 DOI: 10.3390/cimb44120421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by senile plaques formed by amyloid-beta (Aβ) extracellularly and neurofibrillary tangles (NFTs) formed by hyperphosphorylated tau protein intracellularly. Apart from these two features, insulin deficiency and insulin resistance have also been observed in AD brains. Thus, AD has also been referred to as type 3 diabetes by some of the scientists in this field. Insulin plays a pivotal role in learning and memory and is involved in regulating tau phosphorylation though the PI3KAkt-GSK3b signaling pathway. Interestingly, recent studies revealed that in AD brains the microglia transformed into a disease-associated microglia (DAM) status in a TREM2-dependent manner to restrain the toxicity of Aβ and propagation of tau. This also correlated with PI3K-Akt signaling through the adaptor of TREM2. Whether insulin has any effect on microglia activation in AD pathology is unclear so far. However, many studies demonstrated that diabetes increased the risk of AD. In this review, we summarize the main strategies for curing AD, including lowering the level of Aβ, suppressing the phosphorylation of tau, the ablation and/or repopulation of microglia, and especially the supply of insulin. We also propose that attention should be given to the influences of insulin on microglia in AD.
Collapse
|
23
|
Polanco JC, Götz J. Exosomal and vesicle-free tau seeds-propagation and convergence in endolysosomal permeabilization. FEBS J 2022; 289:6891-6907. [PMID: 34092031 DOI: 10.1111/febs.16055] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/19/2021] [Accepted: 06/04/2021] [Indexed: 01/13/2023]
Abstract
In Alzheimer's disease (AD), β-amyloid peptides aggregate to form amyloid plaques, and the microtubule-associated protein tau forms neurofibrillary tangles. However, severity and duration of AD correlate with the stereotypical emergence of tau tangles throughout the brain, suggestive of a gradual region-to-region spreading of pathological tau. The current notion in the field is that misfolded tau seeds propagate transsynaptically and corrupt the proper folding of soluble tau in recipient neurons. This is supported by accumulating evidence showing that in AD, functional connectivity and not proximity predicts the spreading of tau pathology. Tau seeds can be found in two flavors, vesicle-free, that is, naked as in oligomers and fibrils, or encapsulated by membranes of secreted vesicles known as exosomes. Both types of seeds have been shown to propagate between interconnected neurons. Here, we describe potential ways of how their propagation can be controlled in several subcellular compartments by manipulating mechanisms affecting production, neuron-to-neuron transmission, internalization, endosomal escape, and autophagy. We emphasize that although vesicle-free tau seeds and exosomes differ, they share the ability to trigger endolysosomal permeabilization. Such a mechanistic convergence in endolysosomal permeabilization presents itself as a unique opportunity to target both types of tau seeding. We discuss the cellular response to endolysosomal damage that might be key to control permeabilization, and the significant overlap in the seeding mechanism of proteopathic agents other than tau, which suggests that targeting the endolysosomal pathway could pave the way toward developing broad-spectrum treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Juan Carlos Polanco
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
24
|
Kim Y, Park H, Kim Y, Kim SH, Lee JH, Yang H, Kim SJ, Li CM, Lee H, Na DH, Moon S, Shin Y, Kam TI, Lee HW, Kim S, Song JJ, Jung YK. Pathogenic Role of RAGE in Tau Transmission and Memory Deficits. Biol Psychiatry 2022; 93:829-841. [PMID: 36759256 DOI: 10.1016/j.biopsych.2022.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND In tauopathies, brain regions with tau accumulation strongly correlate with clinical symptoms, and spreading of misfolded tau along neural network leads to disease progression. However, the underlying mechanisms by which tau proteins enter neurons during pathological propagation remain unclear. METHODS To identify membrane receptors responsible for neuronal propagation of tau oligomers, we established a cell-based tau uptake assay and screened complementary DNA expression library. Tau uptake and propagation were analyzed in vitro and in vivo using a microfluidic device and stereotactic injection. The cognitive function of mice was assessed using behavioral tests. RESULTS From a genome-wide cell-based functional screening, RAGE (receptor for advanced glycation end products) was isolated to stimulate the cellular uptake of tau oligomers. Rage deficiency reduced neuronal uptake of pathological tau prepared from rTg4510 mouse brains or cerebrospinal fluid from patients with Alzheimer's disease and slowed tau propagation between neurons cultured in a 3-chamber microfluidic device. RAGE levels were increased in the brains of rTg4510 mice and tau oligomer-treated neurons. Rage knockout decreased tau transmission in the brains of nontransgenic mice after injection with Alzheimer's disease patient-derived tau and ameliorated memory loss after injection with GFP-P301L-tau-AAV. Treatment of RAGE antagonist FPS-ZM1 blocked transsynaptic tau propagation and inflammatory responses and alleviated cognitive impairment in rTg4510 mice. CONCLUSIONS These results suggest that in neurons and microglia, RAGE binds to pathological tau and facilitates neuronal tau pathology progression and behavioral deficits in tauopathies.
Collapse
Affiliation(s)
- Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Republic of Korea; School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Park
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Youngwon Kim
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seo-Hyun Kim
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Hoon Lee
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Hanseul Yang
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seo Jin Kim
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cathena Meiling Li
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Haneul Lee
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Do-Hyeong Na
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seowon Moon
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yumi Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Tae-In Kam
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Han-Woong Lee
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Ji-Joon Song
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yong-Keun Jung
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Republic of Korea; School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Odfalk KF, Bieniek KF, Hopp SC. Microglia: Friend and foe in tauopathy. Prog Neurobiol 2022; 216:102306. [PMID: 35714860 PMCID: PMC9378545 DOI: 10.1016/j.pneurobio.2022.102306] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/24/2022] [Accepted: 06/10/2022] [Indexed: 12/16/2022]
Abstract
Aggregation of misfolded microtubule associated protein tau into abnormal intracellular inclusions defines a class of neurodegenerative diseases known as tauopathies. The consistent spatiotemporal progression of tau pathology in Alzheimer's disease (AD) led to the hypothesis that tau aggregates spread in the brain via bioactive tau "seeds" underlying advancing disease course. Recent studies implicate microglia, the resident immune cells of the central nervous system, in both negative and positive regulation of tau pathology. Polymorphisms in genes that alter microglial function are associated with the development of AD and other tauopathies. Experimental manipulation of microglia function can alter tau pathology and microglia-mediated neuroinflammatory cascades can exacerbate tau pathology. Microglia also exert protective functions by mitigating tau spread: microglia internalize tau seeds and have the capacity to degrade them. However, when microglia fail to degrade these tau seeds there are deleterious consequences, including secretion of exosomes containing tau that can spread to neurons. This review explores the intersection of microglia and tau from the perspective of neuropathology, neuroimaging, genetics, transcriptomics, and molecular biology. As tau-targeted therapies such as anti-tau antibodies advance through clinical trials, it is critical to understand the interaction between tau and microglia.
Collapse
Affiliation(s)
- Kristian F Odfalk
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
26
|
Therapeutic Approach to Alzheimer’s Disease: Current Treatments and New Perspectives. Pharmaceutics 2022; 14:pharmaceutics14061117. [PMID: 35745693 PMCID: PMC9228613 DOI: 10.3390/pharmaceutics14061117] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia. The pathophysiology of this disease is characterized by the accumulation of amyloid-β, leading to the formation of senile plaques, and by the intracellular presence of neurofibrillary tangles based on hyperphosphorylated tau protein. In the therapeutic approach to AD, we can identify three important fronts: the approved drugs currently available for the treatment of the disease, which include aducanumab, donepezil, galantamine, rivastigmine, memantine, and a combination of memantine and donepezil; therapies under investigation that work mainly on Aβ pathology and tau pathology, and which include γ-secretase inhibitors, β-secretase inhibitors, α-secretase modulators, aggregation inhibitors, metal interfering drugs, drugs that enhance Aβ clearance, inhibitors of tau protein hyperphosphorylation, tau protein aggregation inhibitors, and drugs that promote the clearance of tau, and finally, other alternative therapies designed to improve lifestyle, thus contributing to the prevention of the disease. Therefore, the aim of this review was to analyze and describe current treatments and possible future alternatives in the therapeutic approach to AD.
Collapse
|
27
|
Kang SS, Meng L, Zhang X, Wu Z, Mancieri A, Xie B, Liu X, Weinshenker D, Peng J, Zhang Z, Ye K. Tau modification by the norepinephrine metabolite DOPEGAL stimulates its pathology and propagation. Nat Struct Mol Biol 2022; 29:292-305. [PMID: 35332321 PMCID: PMC9018606 DOI: 10.1038/s41594-022-00745-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
Abstract
The noradrenergic locus ceruleus (LC) is the first site of detectable tau pathology in Alzheimer's disease (AD), but the mechanisms underlying the selective vulnerability of the LC in AD have not been completely identified. In the present study, we show that DOPEGAL, a monoamine oxidase A (MAO-A) metabolite of norepinephrine (NE), reacts directly with the primary amine on the Lys353 residue of tau to stimulate its aggregation and facilitate its propagation. Inhibition of MAO-A or mutation of the Lys353 residue to arginine (Lys353Arg) decreases tau Lys353-DOPEGAL levels and diminishes tau pathology spreading. Wild-type tau preformed fibrils (PFFs) trigger Lys353-DOPEGAL formation, tau pathology propagation and cognitive impairment in MAPT transgenic mice, all of which are attenuated with PFFs made from the Lys353Arg mutant. Thus, the selective vulnerability of LC neurons in AD may be explained, in part, by NE oxidation via MAO-A into DOPEGAL, which covalently modifies tau and accelerates its aggregation, toxicity and propagation.
Collapse
Affiliation(s)
- Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiping Wu
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ariana Mancieri
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Boer Xie
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, China.
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Shenzhen, China.
| |
Collapse
|
28
|
Hu W, Liu F, Gong CX, Iqbal K. Does proteopathic tau propagate trans-synaptically in the brain? Mol Neurodegener 2022; 17:21. [PMID: 35296344 PMCID: PMC8925219 DOI: 10.1186/s13024-022-00527-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/02/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Wen Hu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA.
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| |
Collapse
|
29
|
Rodrigues PV, de Godoy JVP, Bosque BP, Amorim Neto DP, Tostes K, Palameta S, Garcia-Rosa S, Tonoli CCC, de Carvalho HF, de Castro Fonseca M. Transcellular propagation of fibrillar α-synuclein from enteroendocrine to neuronal cells requires cell-to-cell contact and is Rab35-dependent. Sci Rep 2022; 12:4168. [PMID: 35264710 PMCID: PMC8907230 DOI: 10.1038/s41598-022-08076-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/02/2022] [Indexed: 01/15/2023] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative condition featured by motor dysfunction, death of midbrain dopaminergic neurons and accumulation of α-synuclein (αSyn) aggregates. Growing evidence suggests that PD diagnosis happens late in the disease progression and that the pathology may originate much earlier in the enteric nervous system (ENS) before advancing to the brain, via autonomic fibers. It was recently described that a specific cell type from the gut epithelium named enteroendocrine cells (EECs) possess many neuron-like properties including αSyn expression. By facing the gut lumen and being directly connected with αSyn-containing enteric neurons in a synaptic manner, EECs form a neural circuit between the gastrointestinal tract and the ENS, thereby being a possible key player in the outcome of PD in the gut. We have characterized the progression and the cellular mechanisms involved in αSyn pre-formed fibrils (PFFs) transfer from EECs to neuronal cells. We show that brain organoids efficiently internalize αSyn PFF seeds which triggers the formation of larger intracellular inclusions. In addition, in the enteroendocrine cell line STC-1 and in the neuronal cell line SH-SY5Y, αSyn PFFs induced intracellular calcium (Ca2+) oscillations on an extracellular Ca2+ source-dependent manner and triggered αSyn fibrils internalization by endocytosis. We characterized the spread of αSyn PFFs from enteroendocrine to neuronal cells and showed that this process is dependent on physical cell-to-cell contact and on Rab35 GTPase. Lastly, inhibition of Rab35 increases the clearance of αSyn fibrils by redirecting them to the lysosomal compartment. Therefore, our results reveal mechanisms that contribute to the understanding of how seeded αSyn fibrils promote the progression of αSyn pathology from EECs to neuronal cells shifting the focus of PD etiology to the ENS.
Collapse
Affiliation(s)
- Paulla Vieira Rodrigues
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - João Vitor Pereira de Godoy
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Beatriz Pelegrini Bosque
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Dionísio Pedro Amorim Neto
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Katiane Tostes
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | - Soledad Palameta
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | - Sheila Garcia-Rosa
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | - Celisa Caldana Costa Tonoli
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | | | - Matheus de Castro Fonseca
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil. .,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil. .,Laboratory of Sarkis Mazmanian, Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA, USA.
| |
Collapse
|
30
|
Dujardin S, Fernandes A, Bannon R, Commins C, De Los Santos M, Kamath TV, Hayashi M, Hyman BT. Tau propagation is dependent on the genetic background of mouse strains. Brain Commun 2022; 4:fcac048. [PMID: 35350555 PMCID: PMC8952249 DOI: 10.1093/braincomms/fcac048] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/02/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Progressive cognitive decline in Alzheimer's disease correlates closely with the spread of tau protein aggregation across neural networks of the cortical mantle. We tested the hypothesis that heritable factors may influence the rate of propagation of tau pathology across brain regions in a model system, taking advantage of well-defined genetically diverse background strains in mice. We virally expressed human tau locally in the hippocampus and the entorhinal cortex neurons and monitored the cell-to-cell tau protein spread by immunolabelling. Interestingly, some strains showed more tau spreading than others while tau misfolding accumulated at the same rate in all tested mouse strains. Genetic factors may contribute to tau pathology progression across brain networks, which could help refine mechanisms underlying tau cell-to-cell transfer and accumulation, and potentially provide targets for understanding patient-to-patient variability in the rate of disease progression in Alzheimer's disease.
Collapse
Affiliation(s)
- Simon Dujardin
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Analiese Fernandes
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Riley Bannon
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Caitlin Commins
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Mark De Los Santos
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Tarun V. Kamath
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Bradley T. Hyman
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Ni R. Magnetic Resonance Imaging in Tauopathy Animal Models. Front Aging Neurosci 2022; 13:791679. [PMID: 35145392 PMCID: PMC8821905 DOI: 10.3389/fnagi.2021.791679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
The microtubule-associated protein tau plays an important role in tauopathic diseases such as Alzheimer's disease and primary tauopathies such as progressive supranuclear palsy and corticobasal degeneration. Tauopathy animal models, such as transgenic, knock-in mouse and rat models, recapitulating tauopathy have facilitated the understanding of disease mechanisms. Aberrant accumulation of hyperphosphorylated tau contributes to synaptic deficits, neuroinflammation, and neurodegeneration, leading to cognitive impairment in animal models. Recent advances in molecular imaging using positron emission tomography (PET) and magnetic resonance imaging (MRI) have provided valuable insights into the time course of disease pathophysiology in tauopathy animal models. High-field MRI has been applied for in vivo imaging in animal models of tauopathy, including diffusion tensor imaging for white matter integrity, arterial spin labeling for cerebral blood flow, resting-state functional MRI for functional connectivity, volumetric MRI for neurodegeneration, and MR spectroscopy. In addition, MR contrast agents for non-invasive imaging of tau have been developed recently. Many preclinical MRI indicators offer excellent translational value and provide a blueprint for clinical MRI in the brains of patients with tauopathies. In this review, we summarized the recent advances in using MRI to visualize the pathophysiology of tauopathy in small animals. We discussed the outstanding challenges in brain imaging using MRI in small animals and propose a future outlook for visualizing tau-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Mckean NE, Handley RR, Snell RG. A Review of the Current Mammalian Models of Alzheimer's Disease and Challenges That Need to Be Overcome. Int J Mol Sci 2021; 22:13168. [PMID: 34884970 PMCID: PMC8658123 DOI: 10.3390/ijms222313168] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is one of the looming health crises of the near future. Increasing lifespans and better medical treatment for other conditions mean that the prevalence of this disease is expected to triple by 2050. The impact of AD includes both the large toll on individuals and their families as well as a large financial cost to society. So far, we have no way to prevent, slow, or cure the disease. Current medications can only alleviate some of the symptoms temporarily. Many animal models of AD have been created, with the first transgenic mouse model in 1995. Mouse models have been beset by challenges, and no mouse model fully captures the symptomatology of AD without multiple genetic mutations and/or transgenes, some of which have never been implicated in human AD. Over 25 years later, many mouse models have been given an AD-like disease and then 'cured' in the lab, only for the treatments to fail in clinical trials. This review argues that small animal models are insufficient for modelling complex disorders such as AD. In order to find effective treatments for AD, we need to create large animal models with brains and lifespan that are closer to humans, and underlying genetics that already predispose them to AD-like phenotypes.
Collapse
Affiliation(s)
- Natasha Elizabeth Mckean
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Renee Robyn Handley
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Russell Grant Snell
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
33
|
Meisl G, Hidari E, Allinson K, Rittman T, DeVos SL, Sanchez JS, Xu CK, Duff KE, Johnson KA, Rowe JB, Hyman BT, Knowles TPJ, Klenerman D. In vivo rate-determining steps of tau seed accumulation in Alzheimer's disease. SCIENCE ADVANCES 2021; 7:eabh1448. [PMID: 34714685 PMCID: PMC8555892 DOI: 10.1126/sciadv.abh1448] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 09/09/2021] [Indexed: 05/21/2023]
Abstract
Both the replication of protein aggregates and their spreading throughout the brain are implicated in the progression of Alzheimer’s disease (AD). However, the rates of these processes are unknown and the identity of the rate-determining process in humans has therefore remained elusive. By bringing together chemical kinetics with measurements of tau seeds and aggregates across brain regions, we can quantify their replication rate in human brains. Notably, we obtain comparable rates in several different datasets, with five different methods of tau quantification, from postmortem seed amplification assays to tau PET studies in living individuals. Our results suggest that from Braak stage III onward, local replication, rather than spreading between brain regions, is the main process controlling the overall rate of accumulation of tau in neocortical regions. The number of seeds doubles only every ∼5 years. Thus, limiting local replication likely constitutes the most promising strategy to control tau accumulation during AD.
Collapse
Affiliation(s)
- Georg Meisl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Eric Hidari
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Kieren Allinson
- Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Sarah L. DeVos
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neuro-degenerative Disease, Massachusetts General Hospital, Charlestown, MA 02114, USA
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Justin S. Sanchez
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neuro-degenerative Disease, Massachusetts General Hospital, Charlestown, MA 02114, USA
| | - Catherine K. Xu
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Karen E. Duff
- Dementia Research Institute, University College London, London W1T 7NF, UK
| | - Keith A. Johnson
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neuro-degenerative Disease, Massachusetts General Hospital, Charlestown, MA 02114, USA
| | - James B. Rowe
- Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge CB2 0QQ, UK
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge CB2 7EF, UK
- Cambridge University Hospitals NHS Trust, Cambridge CB2 0SZ, UK
| | - Bradley T. Hyman
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neuro-degenerative Disease, Massachusetts General Hospital, Charlestown, MA 02114, USA
| | - Tuomas P. J. Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- Cavendish Laboratory, University of Cambridge, 19 JJ Thomson Avenue, Cambridge CB3 0HE, UK
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| |
Collapse
|
34
|
Amro Z, Yool AJ, Collins-Praino LE. The potential role of glial cells in driving the prion-like transcellular propagation of tau in tauopathies. Brain Behav Immun Health 2021; 14:100242. [PMID: 34589757 PMCID: PMC8474563 DOI: 10.1016/j.bbih.2021.100242] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
Dementia is one of the leading causes of death worldwide, with tauopathies, a class of diseases defined by pathology associated with the microtubule-enriched protein, tau, as the major contributor. Although tauopathies, such as Alzheimer's disease and Frontotemporal dementia, are common amongst the ageing population, current effective treatment options are scarce, primarily due to the incomplete understanding of disease pathogenesis. The mechanisms via which aggregated forms of tau are able to propagate from one anatomical area to another to cause disease spread and progression is yet unknown. The prion-like hypothesis of tau propagation proposes that tau can propagate along neighbouring anatomical areas in a similar manner to prion proteins in prion diseases, such as Creutzfeldt-Jacob disease. This hypothesis has been supported by a plethora of studies that note the ability of tau to be actively secreted by neurons, propagated and internalised by neighbouring neuronal cells, causing disease spread. Surfacing research suggests a role of reactive astrocytes and microglia in early pre-clinical stages of tauopathy through their inflammatory actions. Furthermore, both glial types are able to internalise and secrete tau from the extracellular space, suggesting a potential role in tau propagation; although understanding the physiological mechanisms by which this can occur remains poorly understood. This review will discuss the current literature around the prion-like propagation of tau, with particular emphasis on glial-mediated neuroinflammation and the contribution it may play in this propagation process.
Collapse
Affiliation(s)
- Zein Amro
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | | |
Collapse
|
35
|
Vogels T, Vargová G, Brezováková V, Quint WH, Hromádka T. Viral Delivery of Non-Mutated Human Truncated Tau to Neurons Recapitulates Key Features of Human Tauopathy in Wild-Type Mice. J Alzheimers Dis 2021; 77:551-568. [PMID: 32675411 DOI: 10.3233/jad-200047] [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] [Indexed: 12/17/2022]
Abstract
BACKGROUND Neuronal accumulation of hyperphosphorylated and truncated tau aggregates is one of the major defining factors and key drivers of neurodegeneration in Alzheimer's disease and other tauopathies. OBJECTIVE We developed an AAV-induced model of tauopathy mediated by human truncated tau protein without familial frontotemporal dementia-related mutations to study tau propagation and the functional consequences of tau pathology. METHODS We performed targeted transductions of the hippocampus or entorhinal cortex in adult mice followed by histological analysis to study the progression of hippocampal tau pathology and tau spreading. We performed behavioral analysis of mice with AAV-induced hippocampal tau pathology. RESULTS AAV-induced hippocampal tau pathology was characterized by tau hyperphosphorylation (AT8 positivity), sarkosyl insolubility, and the presence of neurofibrillary tangles. AAV-induced tau pathology was associated with microgliosis and hypertrophic astrocytes in the absence of cognitive deficits. Additionally, the co-expression of mCherry fluorescent protein and human truncated tau enabled us to detect both local spreading of human tau and spreading from the entorhinal cortex to the synaptically connected dentate gyrus. CONCLUSION Targeted delivery of AAV with truncated tau protein into subcortical and cortical structures of mammalian brains represents an efficient approach for creating temporally and spatially well-defined tau pathology suitable for in vivo studies of tau propagation and neuronal circuit deficits in Alzheimer's disease.
Collapse
Affiliation(s)
- Thomas Vogels
- Axon Neuroscience R & D Services SE, Bratislava, Slovakia
| | - Gréta Vargová
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | - Tomáš Hromádka
- Axon Neuroscience R & D Services SE, Bratislava, Slovakia.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
36
|
Qiu C, Albayram O, Kondo A, Wang B, Kim N, Arai K, Tsai CY, Bassal MA, Herbert MK, Washida K, Angeli P, Kozono S, Stucky JE, Baxley S, Lin YM, Sun Y, Rotenberg A, Caldarone BJ, Bigio EH, Chen X, Tenen DG, Zeidel M, Lo EH, Zhou XZ, Lu KP. Cis P-tau underlies vascular contribution to cognitive impairment and dementia and can be effectively targeted by immunotherapy in mice. Sci Transl Med 2021; 13:13/596/eaaz7615. [PMID: 34078745 DOI: 10.1126/scitranslmed.aaz7615] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 08/14/2020] [Accepted: 03/26/2021] [Indexed: 01/02/2023]
Abstract
Compelling evidence supports vascular contributions to cognitive impairment and dementia (VCID) including Alzheimer's disease (AD), but the underlying pathogenic mechanisms and treatments are not fully understood. Cis P-tau is an early driver of neurodegeneration resulting from traumatic brain injury, but its role in VCID remains unclear. Here, we found robust cis P-tau despite no tau tangles in patients with VCID and in mice modeling key aspects of clinical VCID, likely because of the inhibition of its isomerase Pin1 by DAPK1. Elimination of cis P-tau in VCID mice using cis-targeted immunotherapy, brain-specific Pin1 overexpression, or DAPK1 knockout effectively rescues VCID-like neurodegeneration and cognitive impairment in executive function. Cis mAb also prevents and ameliorates progression of AD-like neurodegeneration and memory loss in mice. Furthermore, single-cell RNA sequencing revealed that young VCID mice display diverse cortical cell type-specific transcriptomic changes resembling old patients with AD, and the vast majority of these global changes were recovered by cis-targeted immunotherapy. Moreover, purified soluble cis P-tau was sufficient to induce progressive neurodegeneration and brain dysfunction by causing axonopathy and conserved transcriptomic signature found in VCID mice and patients with AD with early pathology. Thus, cis P-tau might play a major role in mediating VCID and AD, and antibody targeting it may be useful for early diagnosis, prevention, and treatment of cognitive impairment and dementia after neurovascular insults and in AD.
Collapse
Affiliation(s)
- Chenxi Qiu
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Onder Albayram
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Asami Kondo
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Bin Wang
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nami Kim
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ken Arai
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Cheng-Yu Tsai
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mahmoud A Bassal
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Megan K Herbert
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kazuo Washida
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Peter Angeli
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shingo Kozono
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph E Stucky
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sean Baxley
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yu-Min Lin
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yan Sun
- Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander Rotenberg
- Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Barbara J Caldarone
- NeuroBehavior Laboratory, Harvard NeuroDiscovery Center, Harvard Medical School, Boston, MA 02115, USA
| | - Eileen H Bigio
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiaochun Chen
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Daniel G Tenen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Zeidel
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Eng H Lo
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Xiao Zhen Zhou
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. .,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kun Ping Lu
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. .,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
37
|
Kara E, Crimi A, Wiedmer A, Emmenegger M, Manzoni C, Bandres-Ciga S, D'Sa K, Reynolds RH, Botía JA, Losa M, Lysenko V, Carta M, Heinzer D, Avar M, Chincisan A, Blauwendraat C, García-Ruiz S, Pease D, Mottier L, Carrella A, Beck-Schneider D, Magalhães AD, Aemisegger C, Theocharides APA, Fan Z, Marks JD, Hopp SC, Abramov AY, Lewis PA, Ryten M, Hardy J, Hyman BT, Aguzzi A. An integrated genomic approach to dissect the genetic landscape regulating the cell-to-cell transfer of α-synuclein. Cell Rep 2021; 35:109189. [PMID: 34107263 PMCID: PMC8207177 DOI: 10.1016/j.celrep.2021.109189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/08/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Neuropathological and experimental evidence suggests that the cell-to-cell transfer of α-synuclein has an important role in the pathogenesis of Parkinson's disease (PD). However, the mechanism underlying this phenomenon is not fully understood. We undertook a small interfering RNA (siRNA), genome-wide screen to identify genes regulating the cell-to-cell transfer of α-synuclein. A genetically encoded reporter, GFP-2A-αSynuclein-RFP, suitable for separating donor and recipient cells, was transiently transfected into HEK cells stably overexpressing α-synuclein. We find that 38 genes regulate the transfer of α-synuclein-RFP, one of which is ITGA8, a candidate gene identified through a recent PD genome-wide association study (GWAS). Weighted gene co-expression network analysis (WGCNA) and weighted protein-protein network interaction analysis (WPPNIA) show that those hits cluster in networks that include known PD genes more frequently than expected by random chance. The findings expand our understanding of the mechanism of α-synuclein spread.
Collapse
Affiliation(s)
- Eleanna Kara
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland; Department of Neurodegenerative disease, University College London, London WC1N 3BG, UK
| | - Alessandro Crimi
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Anne Wiedmer
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Marc Emmenegger
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Claudia Manzoni
- Department of Pharmacology, University College London School of Pharmacy, London WC1N 1AX, UK; School of Pharmacy, University of Reading, Reading RG6 6AP, UK
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda, MD 20814, USA
| | - Karishma D'Sa
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Regina H Reynolds
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Juan A Botía
- Department of Neurodegenerative disease, University College London, London WC1N 3BG, UK; Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia 30100, Spain
| | - Marco Losa
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Veronika Lysenko
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Manfredi Carta
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Daniel Heinzer
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Merve Avar
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Andra Chincisan
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | | | - Sonia García-Ruiz
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Daniel Pease
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Lorene Mottier
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Alessandra Carrella
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Dezirae Beck-Schneider
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Andreia D Magalhães
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Caroline Aemisegger
- Center for Microscopy and Image Analysis, University of Zurich, Zurich 8057, Switzerland
| | - Alexandre P A Theocharides
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Zhanyun Fan
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Jordan D Marks
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarah C Hopp
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX 78229, USA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Patrick A Lewis
- Department of Neurodegenerative disease, University College London, London WC1N 3BG, UK; School of Pharmacy, University of Reading, Reading RG6 6AP, UK; Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Mina Ryten
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - John Hardy
- Department of Neurodegenerative disease, University College London, London WC1N 3BG, UK; UK Dementia Research Institute, University College London, London WC1N 3BG, UK; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK; Institute for Advanced Study, the Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland.
| |
Collapse
|
38
|
Robert A, Schöll M, Vogels T. Tau Seeding Mouse Models with Patient Brain-Derived Aggregates. Int J Mol Sci 2021; 22:6132. [PMID: 34200180 PMCID: PMC8201271 DOI: 10.3390/ijms22116132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/23/2022] Open
Abstract
Tauopathies are a heterogeneous class of neurodegenerative diseases characterized by intracellular inclusions of aggregated tau proteins. Tau aggregates in different tauopathies have distinct structural features and can be found in different cell types. Transgenic animal models overexpressing human tau have been used for over two decades in the research of tau pathology. However, these models poorly recapitulate the heterogeneity of tauopathies found in human brains. Recent findings demonstrate that injection of purified tau aggregates from the brains of human tauopathy patients recapitulates both the structural features and cell-type specificity of the tau pathology of the donor tauopathy. These models may therefore have unique translational value in the study of functional consequences of tau pathology, tau-based diagnostics, and tau targeting therapeutics. This review provides an update of the literature relating to seeding-based tauopathy and their potential applications.
Collapse
Affiliation(s)
- Aiko Robert
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London WC1N 3BG, UK; (A.R.); (M.S.)
| | - Michael Schöll
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London WC1N 3BG, UK; (A.R.); (M.S.)
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, 413 45 Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Thomas Vogels
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London WC1N 3BG, UK; (A.R.); (M.S.)
- Department of Psychiatry and Neurochemistry, University of Gothenburg, 413 45 Gothenburg, Sweden
- Sylics (Synaptologics B.V.), 3721 MA Bilthoven, The Netherlands
| |
Collapse
|
39
|
Martin D, Reine F, Herzog L, Igel-Egalon A, Aron N, Michel C, Moudjou M, Fichet G, Quadrio I, Perret-Liaudet A, Andréoletti O, Rezaei H, Béringue V. Prion potentiation after life-long dormancy in mice devoid of PrP. Brain Commun 2021; 3:fcab092. [PMID: 33997785 PMCID: PMC8111064 DOI: 10.1093/braincomms/fcab092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/26/2021] [Accepted: 04/01/2021] [Indexed: 12/26/2022] Open
Abstract
Prions are neurotropic pathogens composed of misfolded assemblies of the host-encoded prion protein PrPC which replicate by recruitment and conversion of further PrPC by an autocatalytic seeding polymerization process. While it has long been shown that mouse-adapted prions cannot replicate and are rapidly cleared in transgenic PrP0/0 mice invalidated for PrPC, these experiments have not been done with other prions, including from natural resources, and more sensitive methods to detect prion biological activity. Using transgenic mice expressing human PrP to bioassay prion infectivity and RT-QuIC cell-free assay to measure prion seeding activity, we report that prions responsible for the most prevalent form of sporadic Creutzfeldt-Jakob disease in human (MM1-sCJD) can persist indefinitely in the brain of intra-cerebrally inoculated PrP0/0 mice. While low levels of seeding activity were measured by RT-QuIC in the brain of the challenged PrP0/0 mice, the bio-indicator humanized mice succumbed at a high attack rate, suggesting relatively high levels of persistent infectivity. Remarkably, these humanized mice succumbed with delayed kinetics as compared to MM1-sCJD prions directly inoculated at low doses, including the limiting one. Yet, the disease that did occur in the humanized mice on primary and subsequent back-passage from PrP0/0 mice shared the neuropathological and molecular characteristics of MM1-sCJD prions, suggesting no apparent strain evolution during lifelong dormancy in PrP0/0 brain. Thus, MM1-sCJD prions can persist for the entire life in PrP0/0 brain with potential disease potentiation on retrotransmission to susceptible hosts. These findings highlight the capacity of prions to persist and rejuvenate in non-replicative environments, interrogate on the type of prion assemblies at work and alert on the risk of indefinite prion persistence with PrP-lowering therapeutic strategies.
Collapse
Affiliation(s)
- Davy Martin
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| | - Fabienne Reine
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| | - Laetitia Herzog
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| | | | - Naima Aron
- INRAE, École Nationale Vétérinaire de Toulouse, IHAP, 31 000 Toulouse, France
| | - Christel Michel
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| | - Mohammed Moudjou
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| | - Guillaume Fichet
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| | - Isabelle Quadrio
- Neurobiology Laboratory, Biochemistry and Molecular Biology Department, Hôpitaux de Lyon, 69 000 Lyon, France.,University of Lyon 1, CNRS UMR5292, INSERM U1028, BioRan, 69 000 Lyon, France
| | - Armand Perret-Liaudet
- Neurobiology Laboratory, Biochemistry and Molecular Biology Department, Hôpitaux de Lyon, 69 000 Lyon, France.,University of Lyon 1, CNRS UMR5292, INSERM U1028, BioRan, 69 000 Lyon, France
| | - Olivier Andréoletti
- INRAE, École Nationale Vétérinaire de Toulouse, IHAP, 31 000 Toulouse, France
| | - Human Rezaei
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| | - Vincent Béringue
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78 350 Jouy-en-Josas, France
| |
Collapse
|
40
|
Continuous Monitoring of Tau-Induced Neurotoxicity in Patient-Derived iPSC-Neurons. J Neurosci 2021; 41:4335-4348. [PMID: 33893219 PMCID: PMC8143197 DOI: 10.1523/jneurosci.2590-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 11/28/2022] Open
Abstract
Tau aggregation within neurons is a critical feature of Alzheimer's disease (AD) and related tauopathies. It is believed that soluble pathologic tau species seed the formation of tau aggregates in a prion-like manner and propagate through connected neurons during the progression of disease. Both soluble and aggregated forms of tau are thought to have neurotoxic properties. In addition, different strains of misfolded tau may cause differential neurotoxicity. In this work, we present an accelerated human neuronal model of tau-induced neurotoxicity that incorporates both soluble tau species and tau aggregation. Using patient-derived induced pluripotent stem cell (iPSC) neurons expressing a tau aggregation biosensor, we develop a cell culture system that allows continuous assessment of both induced tau aggregation and neuronal viability at single-cell resolution for periods of >1 week. We show that exogenous tau “seed” uptake, as measured by tau repeat domain (TauRD) reporter aggregation, increases the risk for subsequent neuronal death in vitro. These results are the first to directly visualize neuronal TauRD aggregation and subsequent cell death in single human iPSC neurons. Specific morphologic strains or patterns of TauRD aggregation are then identified and associated with differing neurotoxicity. Furthermore, we demonstrate that familial AD iPSC neurons expressing the PSEN1 L435F mutation exhibit accelerated TauRD aggregation kinetics and a tau strain propagation bias when compared with control iPSC neurons. SIGNIFICANCE STATEMENT Neuronal intracellular aggregation of the microtubule binding protein tau occurs in Alzheimer's disease and related neurodegenerative tauopathies. Tau aggregates are believed to spread from neuron to neuron via prion-like misfolded tau seeds. Our work develops a human neuronal live-imaging system to visualize seeded tau aggregation and tau-induced neurotoxicity within single neurons. Using an aggregation-sensing tau reporter, we find that neuronal uptake and propagation of tau seeds reduces subsequent survival. In addition, human induced pluripotent stem cell (iPSC) neurons carrying an Alzheimer's disease-causing mutation in presenilin-1 undergo tau seeding more rapidly than control iPSC neurons. However, they do not show subsequent differences in neuronal survival. Finally, specific morphologies of tau aggregates are associated with increased neurotoxicity.
Collapse
|
41
|
Retromer dysfunction at the nexus of tauopathies. Cell Death Differ 2021; 28:884-899. [PMID: 33473181 PMCID: PMC7937680 DOI: 10.1038/s41418-020-00727-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/13/2020] [Accepted: 12/27/2020] [Indexed: 01/30/2023] Open
Abstract
Tauopathies define a broad range of neurodegenerative diseases that encompass pathological aggregation of the microtubule-associated protein tau. Although tau aggregation is a central feature of these diseases, their underlying pathobiology is remarkably heterogeneous at the molecular level. In this review, we summarize critical differences that account for this heterogeneity and contrast the physiological and pathological functions of tau. We focus on the recent understanding of its prion-like behavior that accounts for its spread in the brain. Moreover, we acknowledge the limited appreciation about how upstream cellular changes influence tauopathy. Dysfunction of the highly conserved endosomal trafficking complex retromer is found in numerous tauopathies such as Alzheimer's disease, Pick's disease, and progressive supranuclear palsy, and we discuss how this has emerged as a major contributor to various aspects of neurodegenerative diseases. In particular, we highlight recent investigations that have elucidated the contribution of retromer dysfunction to distinct measures of tauopathy such as tau hyperphosphorylation, aggregation, and impaired cognition and behavior. Finally, we discuss the potential benefit of targeting retromer for modifying disease burden and identify important considerations with such an approach moving toward clinical translation.
Collapse
|
42
|
Asher DM, Belay E, Bigio E, Brandner S, Brubaker SA, Caughey B, Clark B, Damon I, Diamond M, Freund M, Hyman BT, Jucker M, Keene CD, Lieberman AP, Mackiewicz M, Montine TJ, Morgello S, Phelps C, Safar J, Schneider JA, Schonberger LB, Sigurdson C, Silverberg N, Trojanowski JQ, Frosch MP. Risk of Transmissibility From Neurodegenerative Disease-Associated Proteins: Experimental Knowns and Unknowns. J Neuropathol Exp Neurol 2021; 79:1141-1146. [PMID: 33000167 PMCID: PMC7577514 DOI: 10.1093/jnen/nlaa109] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent studies in animal models demonstrate that certain misfolded proteins associated with neurodegenerative diseases can support templated misfolding of cognate native proteins, to propagate across neural systems, and to therefore have some of the properties of classical prion diseases like Creutzfeldt-Jakob disease. The National Institute of Aging convened a meeting to discuss the implications of these observations for research priorities. A summary of the discussion is presented here, with a focus on limitations of current knowledge, highlighting areas that appear to require further investigation in order to guide scientific practice while minimizing potential exposure or risk in the laboratory setting. The committee concluded that, based on all currently available data, although neurodegenerative disease-associated aggregates of several different non-prion proteins can be propagated from humans to experimental animals, there is currently insufficient evidence to suggest more than a negligible risk, if any, of a direct infectious etiology for the human neurodegenerative disorders defined in part by these proteins. Given the importance of this question, the potential for noninvasive human transmission of proteopathic disorders is deserving of further investigation.
Collapse
Affiliation(s)
- David M Asher
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Ermias Belay
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Eileen Bigio
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology Queen Square, London
| | - Scott A Brubaker
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Brychan Clark
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Inger Damon
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Marc Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michelle Freund
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen and German Center for Neurodegenerative Diseases (DZNE), Tübingen
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Miroslaw Mackiewicz
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, California
| | - Susan Morgello
- Departments of Neurology, Neuroscience, and Pathology, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Creighton Phelps
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Jiri Safar
- Departments of Pathology and Neurology, Case Western Reserve University, Cleveland, Ohio
| | - Julie A Schneider
- Department of Neurological Sciences, Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Lawrence B Schonberger
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Christina Sigurdson
- Department of Pathology, University of California - San Diego, San Diego, California
| | - Nina Silverberg
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matthew P Frosch
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, University of Washington, Seattle, Washington.,C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
43
|
Wegmann S, DeVos SL, Zeitler B, Marlen K, Bennett RE, Perez-Rando M, MacKenzie D, Yu Q, Commins C, Bannon RN, Corjuc BT, Chase A, Diez L, Nguyen HOB, Hinkley S, Zhang L, Goodwin A, Ledeboer A, Lam S, Ankoudinova I, Tran H, Scarlott N, Amora R, Surosky R, Miller JC, Robbins AB, Rebar EJ, Urnov FD, Holmes MC, Pooler AM, Riley B, Zhang HS, Hyman BT. Persistent repression of tau in the brain using engineered zinc finger protein transcription factors. SCIENCE ADVANCES 2021; 7:7/12/eabe1611. [PMID: 33741591 PMCID: PMC7978433 DOI: 10.1126/sciadv.abe1611] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/05/2021] [Indexed: 05/12/2023]
Abstract
Neuronal tau reduction confers resilience against β-amyloid and tau-related neurotoxicity in vitro and in vivo. Here, we introduce a novel translational approach to lower expression of the tau gene MAPT at the transcriptional level using gene-silencing zinc finger protein transcription factors (ZFP-TFs). Following a single administration of adeno-associated virus (AAV), either locally into the hippocampus or intravenously to enable whole-brain transduction, we selectively reduced tau messenger RNA and protein by 50 to 80% out to 11 months, the longest time point studied. Sustained tau lowering was achieved without detectable off-target effects, overt histopathological changes, or molecular alterations. Tau reduction with AAV ZFP-TFs was able to rescue neuronal damage around amyloid plaques in a mouse model of Alzheimer's disease (APP/PS1 line). The highly specific, durable, and controlled knockdown of endogenous tau makes AAV-delivered ZFP-TFs a promising approach for the treatment of tau-related human brain diseases.
Collapse
Affiliation(s)
- Susanne Wegmann
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA.
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Sarah L DeVos
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | | | | | - Rachel E Bennett
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Marta Perez-Rando
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Danny MacKenzie
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Qi Yu
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | - Caitlin Commins
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Riley N Bannon
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Bianca T Corjuc
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Alison Chase
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Lisa Diez
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | | | | | - Lei Zhang
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | | | - Stephen Lam
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | - Hung Tran
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | | | | | | | - Ashley B Robbins
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | | | | | | | - Amy M Pooler
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | - Brigit Riley
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | - Bradley T Hyman
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA.
| |
Collapse
|
44
|
Isoform-selective decrease of glycogen synthase kinase-3-beta (GSK-3β) reduces synaptic tau phosphorylation, transcellular spreading, and aggregation. iScience 2021; 24:102058. [PMID: 33554064 PMCID: PMC7848608 DOI: 10.1016/j.isci.2021.102058] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/20/2020] [Accepted: 01/07/2021] [Indexed: 12/27/2022] Open
Abstract
It has been suggested that aberrant activation of glycogen synthase kinase-3-beta (GSK-3β) can trigger abnormal tau hyperphosphorylation and aggregation, which ultimately leads to neuronal/synaptic damage and impaired cognition in Alzheimer disease (AD). We examined if isoform-selective partial reduction of GSK-3β can decrease pathological tau changes, including hyperphosphorylation, aggregation, and spreading, in mice with localized human wild-type tau (hTau) expression in the brain. We used adeno-associated viruses (AAVs) to express hTau locally in the entorhinal cortex of wild-type and GSK-3β hemi-knockout (GSK-3β-HK) mice. GSK-3β-HK mice had significantly less accumulation of hyperphosphorylated tau in synapses and showed a significant decrease of tau protein spread between neurons. In primary neuronal cultures from GSK-3β-HK mice, the aggregation of exogenous FTD-mutant tau was also significantly reduced. These results show that a partial decrease of GSK-3β significantly represses tau-initiated neurodegenerative changes in the brain, and therefore is a promising therapeutic target for AD and other tauopathies. Genetic reduction of GSK-3β decreases synaptic accrual of GSK-3β and p-Tau in mice Reduction of GSK-3β lowers the trans-cellular spread of tau in vivo and in vitro Reduction of GSK-3β diminishes the formation of tau aggregates in vitro
Collapse
|
45
|
Dominguez-Meijide A, Vasili E, Outeiro TF. Pharmacological Modulators of Tau Aggregation and Spreading. Brain Sci 2020; 10:E858. [PMID: 33203009 PMCID: PMC7696562 DOI: 10.3390/brainsci10110858] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022] Open
Abstract
Tauopathies are neurodegenerative disorders characterized by the deposition of aggregates composed of abnormal tau protein in the brain. Additionally, misfolded forms of tau can propagate from cell to cell and throughout the brain. This process is thought to lead to the templated misfolding of the native forms of tau, and thereby, to the formation of newer toxic aggregates, thereby propagating the disease. Therefore, modulation of the processes that lead to tau aggregation and spreading is of utmost importance in the fight against tauopathies. In recent years, several molecules have been developed for the modulation of tau aggregation and spreading. In this review, we discuss the processes of tau aggregation and spreading and highlight selected chemicals developed for the modulation of these processes, their usefulness, and putative mechanisms of action. Ultimately, a stronger understanding of the molecular mechanisms involved, and the properties of the substances developed to modulate them, will lead to the development of safer and better strategies for the treatment of tauopathies.
Collapse
Affiliation(s)
- Antonio Dominguez-Meijide
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073 Goettingen, Germany; (A.D.-M.); (E.V.)
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eftychia Vasili
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073 Goettingen, Germany; (A.D.-M.); (E.V.)
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073 Goettingen, Germany; (A.D.-M.); (E.V.)
- Max Planck Institute for Experimental Medicine, 37075 Goettingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| |
Collapse
|
46
|
Liu M, Sui D, Dexheimer T, Hovde S, Deng X, Wang KW, Lin HL, Chien HT, Kweon HK, Kuo NS, Ayoub CA, Jimenez-Harrison D, Andrews PC, Kwok R, Bochar DA, Kuret J, Fortin J, Tsay YG, Kuo MH. Hyperphosphorylation Renders Tau Prone to Aggregate and to Cause Cell Death. Mol Neurobiol 2020; 57:4704-4719. [PMID: 32780352 PMCID: PMC7530023 DOI: 10.1007/s12035-020-02034-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/22/2020] [Indexed: 01/10/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder without a cure or prevention to date. Hyperphosphorylated tau forms the neurofibrillary tangles (NFTs) that correlate well with the progression of cognitive impairments. Animal studies demonstrated the pathogenic role of hyperphosphorylated tau. Understanding how abnormal phosphorylation renders a normal tau prone to form toxic fibrils is key to delineating molecular pathology and to developing efficacious drugs for AD. Production of a tau bearing the disease-relevant hyperphosphorylation and molecular characters is a pivotal step. Here, we report the preparation and characterization of a recombinant hyperphosphorylated tau (p-tau) with strong relevance to disease. P-tau generated by the PIMAX approach resulted in phosphorylation at multiple epitopes linked to the progression of AD neuropathology. In stark contrast to unmodified tau that required an aggregation inducer, and which had minimal effects on cell functions, p-tau formed inducer-free fibrils that triggered a spike of mitochondrial superoxide, induced apoptosis, and caused cell death at sub-micromolar concentrations. P-tau-induced apoptosis was suppressed by inhibitors for reactive oxygen species. Hyperphosphorylation apparently caused rapid formation of a disease-related conformation. In both aggregation and cytotoxicity, p-tau exhibited seeding activities that converted the unmodified tau into a cytotoxic species with an increased propensity for fibrillization. These characters of p-tau are consistent with the emerging view that hyperphosphorylation causes tau to become an aggregation-prone and cytotoxic species that underlies diffusible pathology in AD and other tauopathies. Our results further suggest that p-tau affords a feasible tool for Alzheimer's disease mechanistic and drug discovery studies.
Collapse
Affiliation(s)
- Mengyu Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
| | - Dexin Sui
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
| | - Thomas Dexheimer
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Stacy Hovde
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
| | - Xiexiong Deng
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
- Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1085, USA
| | - Kuang-Wei Wang
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
| | - Hsin Lian Lin
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, China
| | - Hsiao-Tien Chien
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, China
| | - Hye Kyong Kweon
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Nora Sheen Kuo
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher A Ayoub
- Center for Molecular Neurobiology, Department of Molecular Biochemistry, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Daniela Jimenez-Harrison
- Center for Molecular Neurobiology, Department of Molecular Biochemistry, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Philip C Andrews
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Roland Kwok
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | | | - Jeff Kuret
- Center for Molecular Neurobiology, Department of Molecular Biochemistry, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jessica Fortin
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Yeou-Guang Tsay
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, China
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA.
| |
Collapse
|
47
|
Liu M, Dexheimer T, Sui D, Hovde S, Deng X, Kwok R, Bochar DA, Kuo MH. Hyperphosphorylated tau aggregation and cytotoxicity modulators screen identified prescription drugs linked to Alzheimer's disease and cognitive functions. Sci Rep 2020; 10:16551. [PMID: 33024171 PMCID: PMC7539012 DOI: 10.1038/s41598-020-73680-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
The neurodegenerative Alzheimer's disease (AD) affects more than 30 million people worldwide. There is thus far no cure or prevention for AD. Aggregation of hyperphosphorylated tau in the brain correlates with the cognitive decline of patients of AD and other neurodegenerative tauopathies. Intracerebral injection of tau aggregates isolated from tauopathy brains causes similar pathology in the recipient mice, demonstrating the pathogenic role of abnormally phosphorylated tau. Compounds controlling the aggregation of hyperphosphorylated tau therefore are probable modulators for the disease. Here we report the use of recombinant hyperphosphorylated tau (p-tau) to identify potential tauopathy therapeutics and risk factors. Hyperphosphorylation renders tau prone to aggregate and to impair cell viability. Taking advantage of these two characters of p-tau, we performed a screen of a 1280-compound library, and tested a selective group of prescription drugs in p-tau aggregation and cytotoxicity assays. R-(-)-apomorphine and raloxifene were found to be p-tau aggregation inhibitors that protected p-tau-treated cells. In contrast, a subset of benzodiazepines exacerbated p-tau cytotoxicity apparently via enhancing p-tau aggregation. R-(-)apomorphine and raloxifene have been shown to improve cognition in animals or in humans, whereas benzodiazepines were linked to increased risks of dementia. Our results demonstrate the feasibility and potential of using hyperphosphorylated tau-based assays for AD drug discovery and risk factor identification.
Collapse
Affiliation(s)
- Mengyu Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
| | - Thomas Dexheimer
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Dexin Sui
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
| | - Stacy Hovde
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
| | - Xiexiong Deng
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
- Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1085, USA
| | - Roland Kwok
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | | | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA.
| |
Collapse
|
48
|
Jaunmuktane Z, Brandner S. Invited Review: The role of prion-like mechanisms in neurodegenerative diseases. Neuropathol Appl Neurobiol 2020; 46:522-545. [PMID: 31868945 PMCID: PMC7687189 DOI: 10.1111/nan.12592] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/30/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
The prototype of transmissible neurodegenerative proteinopathies is prion diseases, characterized by aggregation of abnormally folded conformers of the native prion protein. A wealth of mechanisms has been proposed to explain the conformational conversion from physiological protein into misfolded, pathological form, mode of toxicity, propagation from cell-to-cell and regional spread. There is increasing evidence that other neurodegenerative diseases, most notably Alzheimer's disease (Aβ and tau), Parkinson's disease (α-synuclein), frontotemporal dementia (TDP43, tau or FUS) and motor neurone disease (TDP43), exhibit at least some of the misfolded prion protein properties. In this review, we will discuss to what extent each of the properties of misfolded prion protein is known to occur for Aβ, tau, α-synuclein and TDP43, with particular focus on self-propagation through seeding, conformational strains, selective cellular and regional vulnerability, stability and resistance to inactivation, oligomers, toxicity and summarize the most recent literature on transmissibility of neurodegenerative disorders.
Collapse
Affiliation(s)
- Z. Jaunmuktane
- Division of NeuropathologyNational Hospital for Neurology and NeurosurgeryUniversity College London NHS Foundation Trust
- Department of Clinical and Movement Neurosciences and Queen Square Brain Bank for Neurological Disorders
| | - S. Brandner
- Division of NeuropathologyNational Hospital for Neurology and NeurosurgeryUniversity College London NHS Foundation Trust
- Department of Neurodegenerative diseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
| |
Collapse
|
49
|
Goedert M. Tau proteinopathies and the prion concept. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:239-259. [PMID: 32958235 DOI: 10.1016/bs.pmbts.2020.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ordered assembly of a small number of proteins into amyloid filaments is central to age-related neurodegenerative diseases. Tau is the most commonly affected of these proteins. In sporadic diseases, assemblies of tau form in a stochastic manner in certain brain regions, from where they appear to spread in a deterministic way, giving rise to disease symptoms. Over the past decade, multiple lines of evidence have shown that assembled tau behaves like a prion. More recently, electron cryo-microscopy of tau filaments has shown that distinct conformers are present in different diseases, with no inter-individual variation for a given disease.
Collapse
Affiliation(s)
- Michel Goedert
- MRC Laboratory Molecular Biology, Cambridge, United Kingdom.
| |
Collapse
|
50
|
Daude N, Kim C, Kang SG, Eskandari-Sedighi G, Haldiman T, Yang J, Fleck SC, Gomez-Cardona E, Han ZZ, Borrego-Ecija S, Wohlgemuth S, Julien O, Wille H, Molina-Porcel L, Gelpi E, Safar JG, Westaway D. Diverse, evolving conformer populations drive distinct phenotypes in frontotemporal lobar degeneration caused by the same MAPT-P301L mutation. Acta Neuropathol 2020; 139:1045-1070. [PMID: 32219515 PMCID: PMC7244472 DOI: 10.1007/s00401-020-02148-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 01/29/2023]
Abstract
Tau protein accumulation is a common denominator of major dementias, but this process is inhomogeneous, even when triggered by the same germline mutation. We considered stochastic misfolding of human tau conformers followed by templated conversion of native monomers as an underlying mechanism and derived sensitive conformational assays to test this concept. Assessments of brains from aged TgTauP301L transgenic mice revealed a prodromal state and three distinct signatures for misfolded tau. Frontotemporal lobar degeneration (FTLD)-MAPT-P301L patients with different clinical phenotypes also displayed three signatures, two resembling those found in TgTauP301L mice. As physicochemical and cell bioassays confirmed diverse tau strains in the mouse and human brain series, we conclude that evolution of diverse tau conformers is intrinsic to the pathogenesis of this uni-allelic form of tauopathy. In turn, effective therapeutic interventions in FTLD will need to address evolving repertoires of misfolded tau species rather than singular, static molecular targets.
Collapse
Affiliation(s)
- Nathalie Daude
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
| | - Chae Kim
- Department of Pathology, Case Western Reserve University, Institute of Pathology Building, Rm 406, 2085 Adelbert Road, Cleveland, OH, 44106-4907, USA
| | - Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
| | - Ghazaleh Eskandari-Sedighi
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Tracy Haldiman
- Department of Pathology, Case Western Reserve University, Institute of Pathology Building, Rm 406, 2085 Adelbert Road, Cleveland, OH, 44106-4907, USA
| | - Jing Yang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
| | - Shelaine C Fleck
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Zhuang Zhuang Han
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Sergi Borrego-Ecija
- Neurological Tissue Bank of the Biobanc, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Serene Wohlgemuth
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Holger Wille
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc, Hospital Clinic, IDIBAPS, Barcelona, Spain
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Jiri G Safar
- Department of Pathology, Case Western Reserve University, Institute of Pathology Building, Rm 406, 2085 Adelbert Road, Cleveland, OH, 44106-4907, USA.
- Department of Neurology, Case Western Reserve University, Institute of Pathology Building, Rm 406, 2085 Adelbert Road, Cleveland, OH, 44106-4907, USA.
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, T6G 2M8, Canada.
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|