22,415 results on '"tauopathies"'
Search Results
2. Proteopathic seed amplification assays in easily accessible specimens for human synucleinopathies, tauopathies, and prionopathies: A scoping review
- Author
-
Salciccia, Clara, Costanzo, Matteo, Ruocco, Giulia, Porreca, Flavia, Vivacqua, Giorgio, Fabbrini, Giovanni, Belvisi, Daniele, Ladogana, Anna, and Poleggi, Anna
- Published
- 2025
- Full Text
- View/download PDF
3. Pragmatic algorithm for visual assessment of 4-Repeat tauopathies in [18F]PI-2620 PET Scans
- Author
-
Bauer, Theresa, Brendel, Matthias, Zaganjori, Mirlind, Bernhardt, Alexander M., Jäck, Alexander, Stöcklein, Sophia, Scheifele, Maximilian, Levin, Johannes, van Eimeren, Thilo, Drzezga, Alexander, Sabri, Osama, Barthel, Henryk, Perneczky, Robert, Höglinger, Günter, Franzmeier, Nicolai, and Gnörich, Johannes
- Published
- 2025
- Full Text
- View/download PDF
4. Effects of blood-brain barrier opening using ultrasound on tauopathies: A systematic review
- Author
-
Géraudie, Amandine, De Rossi, Pierre, Canney, Michael, Carpentier, Alexandre, and Delatour, Benoît
- Published
- 2025
- Full Text
- View/download PDF
5. A tau dephosphorylation-targeting chimera selectively recruits protein phosphatase-1 to ameliorate Alzheimer’s disease and tauopathies
- Author
-
Xiao, Yue, Wei, Linyu, Su, Jingfen, Lei, Huiyang, Sun, Fei, Li, Mengzhu, Li, Shihong, Wang, Xiaochuan, Zheng, Jie, and Wang, Jian-Zhi
- Published
- 2024
- Full Text
- View/download PDF
6. Atypical Parkinsonian Disorders
- Author
-
Olfati, Nahid, Akhoundi, Fahimeh H., and Litvan, Irene
- Published
- 2024
- Full Text
- View/download PDF
7. Botulinum neurotoxin A modulates the axonal release of pathological tau in hippocampal neurons
- Author
-
Panzi, Chiara, Surana, Sunaina, De La-Rocque, Samantha, Moretto, Edoardo, Lazo, Oscar Marcelo, and Schiavo, Giampietro
- Published
- 2023
- Full Text
- View/download PDF
8. Searching for the cellular underpinnings of the selective vulnerability to tauopathic insults in Alzheimer’s disease
- Author
-
Torok, Justin, Maia, Pedro D, Anand, Chaitali, and Raj, Ashish
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Aging ,Genetics ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Alzheimer's Disease ,Brain Disorders ,Acquired Cognitive Impairment ,Neurodegenerative ,Dementia ,Neurosciences ,2.1 Biological and endogenous factors ,Neurological ,Animals ,Alzheimer Disease ,Mice ,tau Proteins ,Tauopathies ,Mice ,Transgenic ,Disease Models ,Animal ,Neurons ,Hippocampus ,Male ,Humans ,Biological sciences ,Biomedical and clinical sciences - Abstract
Neurodegenerative diseases such as Alzheimer's disease exhibit pathological changes in the brain that proceed in a stereotyped and regionally specific fashion. However, the cellular underpinnings of regional vulnerability are poorly understood, in part because whole-brain maps of a comprehensive collection of cell types have been inaccessible. Here, we deployed a recent cell-type mapping pipeline, Matrix Inversion and Subset Selection (MISS), to determine the brain-wide distributions of pan-hippocampal and neocortical cells in the mouse, and then used these maps to identify general principles of cell-type-based selective vulnerability in PS19 mouse models. We found that hippocampal glutamatergic neurons as a whole were significantly positively associated with regional tau deposition, suggesting vulnerability, while cortical glutamatergic and GABAergic neurons were negatively associated. We also identified oligodendrocytes as the single-most strongly negatively associated cell type. Further, cell-type distributions were more predictive of end-time-point tau pathology than AD-risk-gene expression. Using gene ontology analysis, we found that the genes that are directly correlated to tau pathology are functionally distinct from those that constitutively embody the vulnerable cells. In short, we have elucidated cell-type correlates of tau deposition across mouse models of tauopathy, advancing our understanding of selective cellular vulnerability at a whole-brain level.
- Published
- 2025
9. APOE Christchurch enhances a disease-associated microglial response to plaque but suppresses response to tau pathology
- Author
-
Tran, Kristine M, Kwang, Nellie E, Butler, Claire A, Gomez-Arboledas, Angela, Kawauchi, Shimako, Mar, Cassandra, Chao, Donna, Barahona, Rocio A, Da Cunha, Celia, Tsourmas, Kate I, Shi, Zechuan, Wang, Shuling, Collins, Sherilyn, Walker, Amber, Shi, Kai-Xuan, Alcantara, Joshua A, Neumann, Jonathan, Duong, Duc M, Seyfried, Nicholas T, Tenner, Andrea J, LaFerla, Frank M, Hohsfield, Lindsay A, Swarup, Vivek, MacGregor, Grant R, and Green, Kim N
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Aging ,Alzheimer's Disease ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Acquired Cognitive Impairment ,Genetics ,Neurodegenerative ,Neurosciences ,Dementia ,Brain Disorders ,2.1 Biological and endogenous factors ,Neurological ,Animals ,Microglia ,Mice ,Alzheimer Disease ,Plaque ,Amyloid ,tau Proteins ,Mice ,Transgenic ,Tauopathies ,Disease Models ,Animal ,Apolipoproteins E ,Humans ,Brain ,APOE Christchurch ,PS19 ,5xFAD ,DAM ,Amyloid ,Tau ,Resilience ,Clinical Sciences ,Neurology & Neurosurgery ,Biochemistry and cell biology - Abstract
BackgroundApolipoprotein E ε4 (APOE4) is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD). A recent case report identified a rare variant in APOE, APOE3-R136S (Christchurch), proposed to confer resistance to autosomal dominant Alzheimer's Disease (AD). However, it remains unclear whether and how this variant exerts its protective effects.MethodsWe introduced the R136S variant into mouse Apoe (ApoeCh) and investigated its effect on the development of AD-related pathology using the 5xFAD model of amyloidosis and the PS19 model of tauopathy. We used immunohistochemical and biochemical analysis along with single-cell spatial omics and bulk proteomics to explore the impact of the ApoeCh variant on AD pathological development and the brain's response to plaques and tau.ResultsIn 5xFAD mice, ApoeCh enhances a Disease-Associated Microglia (DAM) phenotype in microglia surrounding plaques, and reduces plaque load, dystrophic neurites, and plasma neurofilament light chain. By contrast, in PS19 mice, ApoeCh suppresses the microglial and astrocytic responses to tau-laden neurons and does not reduce tau accumulation or phosphorylation, but partially rescues tau-induced synaptic and myelin loss. We compared how microglia responses differ between the two mouse models to elucidate the distinct DAM signatures induced by ApoeCh. We identified upregulation of antigen presentation-related genes in the DAM response in a PS19 compared to a 5xFAD background, suggesting a differential response to amyloid versus tau pathology that is modulated by the presence of ApoeCh. Bulk proteomics show upregulated mitochondrial protein abundance with ApoeCh in 5xFAD mice, but reductions in mitochondrial and translation associated proteins in PS19 mice.ConclusionsThese findings highlight the ability of the ApoeCh variant to modulate microglial responses based on the type of pathology, enhancing DAM reactivity in amyloid models and dampening neuroinflammation to promote protection in tau models. This suggests that the Christchurch variant's protective effects likely involve multiple mechanisms, including changes in receptor binding and microglial programming.
- Published
- 2025
10. A Study of XPro1595 in Patients with Early Alzheimer's Disease with Biomarkers of Inflammation (MINDFuL)
- Published
- 2025
11. A Study of Donanemab (LY3002813) in Participants With Early Symptomatic Alzheimer's Disease (TRAILBLAZER-ALZ 5)
- Published
- 2025
12. Tau PET/CT in Various Tau-Related Disease Patients
- Author
-
Tianjin Medical University General Hospital and Shaobo Yao, PhD, Prof.
- Published
- 2024
13. A framework for translating tauopathy therapeutics: Drug discovery to clinical trials
- Author
-
Feldman, Howard H, Cummings, Jeffrey L, Boxer, Adam L, Staffaroni, Adam M, Knopman, David S, Rizzo, Stacey J Sukoff, Territo, Paul R, Arnold, Steven E, Ballard, Clive, Beher, Dirk, Boeve, Bradley F, Dacks, Penny A, Diaz, Kristophe, Ewen, Colin, Fiske, Brian, Gonzalez, M Isabel, Harris, Glenn A, Hoffman, Beth J, Martinez, Terina N, McDade, Eric, Nisenbaum, Laura K, Palma, Jose‐Alberto, Quintana, Melanie, Rabinovici, Gil D, Rohrer, Jonathan D, Rosen, Howard J, Troyer, Matthew D, Kim, Doo Yeon, Tanzi, Rudolph E, Zetterberg, Henrik, Ziogas, Nick K, May, Patrick C, and Rommel, Amy
- Subjects
Biomedical and Clinical Sciences ,Biological Psychology ,Clinical Sciences ,Neurosciences ,Psychology ,Acquired Cognitive Impairment ,Frontotemporal Dementia (FTD) ,Aging ,Rare Diseases ,Alzheimer's Disease ,Dementia ,Neurodegenerative ,Brain Disorders ,Clinical Research ,Orphan Drug ,Alzheimer's Disease Related Dementias (ADRD) ,Clinical Trials and Supportive Activities ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,5.1 Pharmaceuticals ,Neurological ,Humans ,Tauopathies ,Drug Discovery ,Clinical Trials as Topic ,Biomarkers ,Translational Research ,Biomedical ,tau Proteins ,Drug Development ,biomarkers ,development ,early-phase clinical trials ,preclinical ,tauopathy ,therapeutics ,early‐phase clinical trials ,Geriatrics ,Clinical sciences ,Biological psychology - 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.
- Published
- 2024
14. CW2IP2: Imaging and Diagnostic Assessments
- Author
-
Ilya Nasrallah, Principal Investigator
- Published
- 2024
15. Establishing 18F PMPBB3 (APN 1607) PET Imaging Markers for Diagnosis of Tauopathy Parkinsonism Syndromes
- Published
- 2024
16. Neurodegenerative Diseases Progression Markers (MARKERS-NDD) (MARKERS-NDD)
- Author
-
IRCCS San Raffaele Roma, San Raffaele Telematic University, University of Urbino 'Carlo Bo', University of Cassino and Southern Lazio, University of Roma La Sapienza, and Politecnico di Milano
- Published
- 2024
17. TMEM16F exacerbates tau pathology and mediates phosphatidylserine exposure in phospho-tau-burdened neurons.
- Author
-
Zubia, Mario, Yong, Adeline, Holtz, Kristen, Huang, Eric, Jan, Yuh, and Jan, Lily
- Subjects
P301S (PS19) ,TMEM16F ,lipid scrambling ,phosphatidylserine exposure ,tauopathy ,Animals ,Anoctamins ,Phosphatidylserines ,Neurons ,tau Proteins ,Mice ,Tauopathies ,Humans ,Microglia ,Phosphorylation ,Mice ,Transgenic ,Disease Models ,Animal ,Phospholipid Transfer Proteins ,Brain ,Mice ,Knockout - Abstract
TMEM16F is a calcium-activated phospholipid scramblase and nonselective ion channel, which allows the movement of lipids bidirectionally across the plasma membrane. While the functions of TMEM16F have been extensively characterized in multiple cell types, the role of TMEM16F in the central nervous system remains largely unknown. Here, we sought to study how TMEM16F in the brain may be involved in neurodegeneration. Using a mouse model that expresses the pathological P301S human tau (PS19 mouse), we found reduced tauopathy and microgliosis in 6- to 7-mo-old PS19 mice lacking TMEM16F. Furthermore, this reduction of pathology can be recapitulated in the PS19 mice with TMEM16F removed from neurons, while removal of TMEM16F from microglia of PS19 mice did not significantly impact tauopathy at this time point. Moreover, TMEM16F mediated aberrant phosphatidylserine exposure in neurons with phospho-tau burden. These studies raise the prospect of targeting TMEM16F in neurons as a potential treatment of neurodegeneration.
- Published
- 2024
18. Anti-acetylated-tau immunotherapy is neuroprotective in tauopathy and brain injury.
- Author
-
Parra Bravo, Celeste, Krukowski, Karen, Barker, Sarah, Wang, Chao, Li, Yaqiao, Fan, Li, Vázquez-Rosa, Edwin, Shin, Min-Kyoo, Wong, Man, McCullough, Louise, Kitagawa, Ryan, Choi, H, Cacace, Angela, Sinha, Subhash, Pieper, Andrew, Rosi, Susanna, Chen, Xu, and Gan, Li
- Subjects
Acetylated tau ,Human plasma ,Immunotherapy ,TBI ,Tauopathy ,Animals ,Tauopathies ,tau Proteins ,Mice ,Acetylation ,Humans ,Immunotherapy ,Disease Models ,Animal ,Mice ,Transgenic ,Brain Injuries ,Traumatic ,Brain Injuries ,Brain ,Neuroprotective Agents - Abstract
BACKGROUND: Tau is aberrantly acetylated in various neurodegenerative conditions, including Alzheimers disease, frontotemporal lobar degeneration (FTLD), and traumatic brain injury (TBI). Previously, we reported that reducing acetylated tau by pharmacologically inhibiting p300-mediated tau acetylation at lysine 174 reduces tau pathology and improves cognitive function in animal models. METHODS: We investigated the therapeutic efficacy of two different antibodies that specifically target acetylated lysine 174 on tau (ac-tauK174). We treated PS19 mice, which harbor the P301S tauopathy mutation that causes FTLD, with anti-ac-tauK174 and measured effects on tau pathology, neurodegeneration, and neurobehavioral outcomes. Furthermore, PS19 mice received treatment post-TBI to evaluate the ability of the immunotherapy to prevent TBI-induced exacerbation of tauopathy phenotypes. Ac-tauK174 measurements in human plasma following TBI were also collected to establish a link between trauma and acetylated tau levels, and single nuclei RNA-sequencing of post-TBI brain tissues from treated mice provided insights into the molecular mechanisms underlying the observed treatment effects. RESULTS: Anti-ac-tauK174 treatment mitigates neurobehavioral impairment and reduces tau pathology in PS19 mice. Ac-tauK174 increases significantly in human plasma 24 h after TBI, and anti-ac-tauK174 treatment of PS19 mice blocked TBI-induced neurodegeneration and preserved memory functions. Anti-ac-tauK174 treatment rescues alterations of microglial and oligodendrocyte transcriptomic states following TBI in PS19 mice. CONCLUSIONS: The ability of anti-ac-tauK174 treatment to rescue neurobehavioral impairment, reduce tau pathology, and rescue glial responses demonstrates that targeting tau acetylation at K174 is a promising neuroprotective therapeutic approach to human tauopathies resulting from TBI or genetic disease.
- Published
- 2024
19. Precision proteoform design for 4R tau isoform selective templated aggregation
- Author
-
Longhini, Andrew P, DuBose, Austin, Lobo, Samuel, Vijayan, Vishnu, Bai, Yeran, Rivera, Erica Keane, Sala-Jarque, Julia, Nikitina, Arina, Carrettiero, Daniel C, Unger, Matthew T, Sclafani, Olivia R, Fu, Valerie, Beckett, Emily R, Vigers, Michael, Buée, Luc, Landrieu, Isabelle, Shell, Scott, Shea, Joan E, Han, Songi, and Kosik, Kenneth S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Aging ,Brain Disorders ,Neurosciences ,Alzheimer's Disease ,Alzheimer's Disease Related Dementias (ADRD) ,Rare Diseases ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Neurodegenerative ,Dementia ,Frontotemporal Dementia (FTD) ,Acquired Cognitive Impairment ,Humans ,tau Proteins ,Tauopathies ,Protein Isoforms ,Prions ,Peptides ,Amino Acids ,amyloidogenic core ,prion-like templating ,protein misfolding ,tauopathies - Abstract
Prion-like spread of disease-specific tau conformers is a hallmark of all tauopathies. A 19-residue probe peptide containing a P301L mutation and spanning the R2/R3 splice junction of tau folds and stacks into seeding-competent fibrils and induces aggregation of 4R, but not 3R tau. These tau peptide fibrils propagate aggregated intracellular tau over multiple generations, have a high β-sheet content, a colocalized lipid signal, and adopt a well-defined U-shaped fold found in 4R tauopathy brain-derived fibrils. Fully atomistic replica exchange molecular dynamics (MD) simulations were used to compute the free energy landscapes of the conformational ensemble of the peptide monomers. These identified an aggregation-prohibiting β-hairpin structure and an aggregation-competent U-fold unique to 4R tauopathy fibrils. Guided by MD simulations, we identified that the N-terminal-flanking residues to PHF6, which slightly vary between 4R and 3R isoforms, modulate seeding. Strikingly, when a single amino acid switch at position 305 replaced the serine of 4R tau with a lysine from the corresponding position in the first repeat of 3R tau, the seeding induced by the 19-residue peptide was markedly reduced. Conversely, a 4R tau mimic with three repeats, prepared by replacing those amino acids in the first repeat with those amino acids uniquely present in the second repeat, recovered aggregation when exposed to the 19-residue peptide. These peptide fibrils function as partial prions to recruit naive 4R tau-ten times the length of the peptide-and serve as a critical template for 4R tauopathy propagation. These results hint at opportunities for tau isoform-specific therapeutic interventions.
- Published
- 2024
20. Prominent tauopathy and intracellular β-amyloid accumulation triggered by genetic deletion of cathepsin D: implications for Alzheimer disease pathogenesis.
- Author
-
Terron, Heather, Parikh, Sagar, Abdul-Hay, Samer, Sahara, Tomoko, Kang, Dongcheul, Dickson, Dennis, Saftig, Paul, LaFerla, Frank, Lane, Shelley, and Leissring, Malcolm
- Subjects
Alzheimer disease ,Amyloid-β protein ,Cathepsin D ,Lysosomes ,Neurofibrillary tangles ,Tauopathy ,Aged ,Animals ,Humans ,Mice ,Alzheimer Disease ,Amyloid beta-Peptides ,Amyloid beta-Protein Precursor ,Cathepsin D ,Disease Models ,Animal ,Mice ,Knockout ,Mice ,Transgenic ,tau Proteins ,Tauopathies - Abstract
BACKGROUND: Cathepsin D (CatD) is a lysosomal protease that degrades both the amyloid-β protein (Aβ) and the microtubule-associated protein, tau, which accumulate pathognomonically in Alzheimer disease (AD), but few studies have examined the role of CatD in the development of Aβ pathology and tauopathy in vivo. METHODS: CatD knockout (KO) mice were crossed to human amyloid precursor protein (hAPP) transgenic mice, and amyloid burden was quantified by ELISA and immunohistochemistry (IHC). Tauopathy in CatD-KO mice, as initially suggested by Gallyas silver staining, was further characterized by extensive IHC and biochemical analyses. Controls included human tau transgenic mice (JNPL3) and another mouse model of a disease (Krabbe A) characterized by pronounced lysosomal dysfunction. Additional experiments examined the effects of CatD inhibition on tau catabolism in vitro and in cultured neuroblastoma cells with inducible expression of human tau. RESULTS: Deletion of CatD in hAPP transgenic mice triggers large increases in cerebral Aβ, manifesting as intense, exclusively intracellular aggregates; extracellular Aβ deposition, by contrast, is neither triggered by CatD deletion, nor affected in older, haploinsufficient mice. Unexpectedly, CatD-KO mice were found to develop prominent tauopathy by just ∼ 3 weeks of age, accumulating sarkosyl-insoluble, hyperphosphorylated tau exceeding the pathology present in aged JNPL3 mice. CatD-KO mice exhibit pronounced perinuclear Gallyas silver staining reminiscent of mature neurofibrillary tangles in human AD, together with widespread phospho-tau immunoreactivity. Striking increases in sarkosyl-insoluble phospho-tau (∼ 1250%) are present in CatD-KO mice but notably absent from Krabbe A mice collected at an identical antemortem interval. In vitro and in cultured cells, we show that tau catabolism is slowed by blockade of CatD proteolytic activity, including via competitive inhibition by Aβ42. CONCLUSIONS: Our findings support a major role for CatD in the proteostasis of both Aβ and tau in vivo. To our knowledge, the CatD-KO mouse line is the only model to develop detectable Aβ accumulation and profound tauopathy in the absence of overexpression of hAPP or human tau with disease-associated mutations. Given that tauopathy emerges from disruption of CatD, which can itself be potently inhibited by Aβ42, our findings suggest that impaired CatD activity may represent a key mechanism linking amyloid accumulation and tauopathy in AD.
- Published
- 2024
21. Mathematical topology and geometry-based classification of tauopathies.
- Author
-
Sugiyama, Masumi, Kosik, Kenneth, and Panagiotou, Eleni
- Subjects
Humans ,Alzheimer Disease ,Cryoelectron Microscopy ,Tauopathies ,tau Proteins ,Supranuclear Palsy ,Progressive ,Cerebral Cortex ,Brain - Abstract
Neurodegenerative diseases, like Alzheimers, are associated with the presence of neurofibrillary lesions formed by tau protein filaments in the cerebral cortex. While it is known that different morphologies of tau filaments characterize different neurodegenerative diseases, there are few metrics of global and local structure complexity that enable to quantify their structural diversity rigorously. In this manuscript, we employ for the first time mathematical topology and geometry to classify neurodegenerative diseases by using cryo-electron microscopy structures of tau filaments that are available in the Protein Data Bank. By employing mathematical topology metrics (Gauss linking integral, writhe and second Vassiliev measure) we achieve a consistent, but more refined classification of tauopathies, than what was previously observed through visual inspection. Our results reveal a hierarchy of classification from global to local topology and geometry characteristics. In particular, we find that tauopathies can be classified with respect to the handedness of their global conformations and the handedness of the relative orientations of their repeats. Progressive supranuclear palsy is identified as an outlier, with a more complex structure than the rest, reflected by a small, but observable knotoid structure (a diagrammatic structure representing non-trivial topology). This topological characteristic can be attributed to a pattern in the beginning of the R3 repeat that is present in all tauopathies but at different extent. Moreover, by comparing single filament to paired filament structures within tauopathies we find a consistent change in the side-chain orientations with respect to the alpha carbon atoms at the area of interaction.
- Published
- 2024
22. Disentangling tau: One protein, many therapeutic approaches
- Author
-
Lane-Donovan, Courtney and Boxer, Adam L
- Subjects
Biomedical and Clinical Sciences ,Biological Psychology ,Neurosciences ,Psychology ,Pharmacology and Pharmaceutical Sciences ,Brain Disorders ,Alzheimer's Disease Related Dementias (ADRD) ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Alzheimer's Disease ,Dementia ,Rare Diseases ,Acquired Cognitive Impairment ,Aging ,Neurodegenerative ,Frontotemporal Dementia (FTD) ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,Neurological ,Humans ,tau Proteins ,Alzheimer Disease ,Tauopathies ,Supranuclear Palsy ,Progressive ,Neurodegenerative Diseases ,Tau ,Neurodegeneration ,Alzheimer's disease ,Progressive supranuclear palsy ,Immunotherapy ,Clinical trials ,Public Health and Health Services ,Neurology & Neurosurgery ,Pharmacology and pharmaceutical sciences ,Biological psychology - Abstract
The tauopathies encompass over 20 adult neurodegenerative diseases and are characterized by the dysfunction and accumulation of insoluble tau protein. Among them, Alzheimer's disease, frontotemporal dementia, and progressive supranuclear palsy collectively impact millions of patients and their families worldwide. Despite years of drug development using a variety of mechanisms of action, no therapeutic directed against tau has been approved for clinical use. This raises important questions about our current model of tau pathology and invites thoughtful consideration of our approach to nonclinical models and clinical trial design. In this article, we review what is known about the biology and genetics of tau, placing it in the context of current and failed clinical trials. We highlight potential reasons for the lack of success to date and offer suggestions for new pathways in therapeutic development. Overall, our viewpoint to the future is optimistic for this important group of neurodegenerative diseases.
- Published
- 2024
23. A new paradigm for neurodegenerative diseases classification: A clinical perspective
- Author
-
Giannakis, Alexandros and Konitsiotis, Spiridon
- Published
- 2025
- Full Text
- View/download PDF
24. An Open-Label Extension of XPro1595 in Patients With Alzheimer's Disease
- Published
- 2024
25. A Study to Evaluate the Safety, Tolerability and Immunogenicity of Tau Targeted Vaccines in Participants With Early Alzheimer's Disease
- Author
-
Janssen Research & Development, LLC
- Published
- 2024
26. Study of Biodistribution, Metabolism, Excretion and Brain Uptake11C-M503
- Published
- 2024
27. Study of Biodistribution, Metabolism, Excretion and Brain Uptake of 11C-HY-2-15
- Published
- 2024
28. Synaptic sabotage: How Tau and α-Synuclein undermine synaptic health.
- Author
-
Uytterhoeven, Valerie, Verstreken, Patrik, and Nachman, Eliana
- Subjects
- *
ALPHA-synuclein , *TAU proteins , *ALZHEIMER'S disease , *PARKINSON'S disease , *TAUOPATHIES - Abstract
Synaptic dysfunction is one of the earliest cellular defects observed in Alzheimer's disease (AD) and Parkinson's disease (PD), occurring before widespread protein aggregation, neuronal loss, and cognitive decline. While the field has focused on the aggregation of Tau and α-Synuclein (α-Syn), emerging evidence suggests that these proteins may drive presynaptic pathology even before their aggregation. Therefore, understanding the mechanisms by which Tau and α-Syn affect presynaptic terminals offers an opportunity for developing innovative therapeutics aimed at preserving synapses and potentially halting neurodegeneration. This review focuses on the molecular defects that converge on presynaptic dysfunction caused by Tau and α-Syn. Both proteins have physiological roles in synapses. However, during disease, they acquire abnormal functions due to aberrant interactions and mislocalization. We provide an overview of current research on different essential presynaptic pathways influenced by Tau and α-Syn. Finally, we highlight promising therapeutic targets aimed at maintaining synaptic function in both tauopathies and synucleinopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
29. Hippocampal atrophy over two years in relation to tau, amyloid-β and memory in older adults.
- Author
-
Aumont, Etienne, Bedard, Marc-André, Bussy, Aurélie, Arias, Jaime Fernandez, Tissot, Cecile, Hall, Brandon J., Therriault, Joseph, Rahmouni, Nesrine, Stevenson, Jenna, Servaes, Stijn, Macedo, Arthur C., Vitali, Paolo, Poltronetti, Nina Margherita, Fliaguine, Olga, Trudel, Lydia, Gauthier, Serge, Chakravarty, Mallar M., and Rosa-Neto, Pedro
- Subjects
- *
HIPPOCAMPUS (Brain) , *ALZHEIMER'S disease , *ENTORHINAL cortex , *CEREBRAL atrophy , *TAUOPATHIES - Abstract
In this longitudinal brain imaging study, we aimed to characterize hippocampal tau accumulation and subfield atrophy relative to cortical amyloid-β and memory performance. We measured tau-PET in regions associated with Braak stages I to VI, global amyloid-PET burden, hippocampal subfield volumes and memory assessments from 173 participants aged 55–85. Eighty-six of these participants were tested again two years later. Tau-PET change in the Braak II region, corresponding to the hippocampus and the entorhinal cortex, was significantly associated with the cornu ammonis 1 (CA1) atrophy and memory score. This CA1 atrophy did not significantly mediate the association between tau and memory, nor did global amyloid-PET burden correlate with tau-PET changes in the Braak II region. Longitudinal hippocampal tau accumulation is amyloid-β-independent and co-localized with subfield atrophy. As tau-associated memory decline seems to be independent from hippocampal atrophy, other mechanisms could contribute to the deficit. • Hippocampal and entorhinal tau accumulation is amyloid-β-independent. • Baseline tau load is not specific to local longitudinal hippocampal atrophy. • CA1 and subiculum atrophy mirror longitudinal hippocampal tau pathology. • Challenge the hippocampal volume's mediation of the tau-memory link. • Memory decline is tied to tau beyond hippocampal subfields atrophy. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
30. Understanding retinal tau pathology through functional 2D and 3D iPSC-derived in vitro retinal models.
- Author
-
Mautone, Lorenza, Cordella, Federica, Soloperto, Alessandro, Ghirga, Silvia, Di Gennaro, Giorgia, Gigante, Ylenia, and Di Angelantonio, Silvia
- Subjects
- *
INDUCED pluripotent stem cells , *PLURIPOTENT stem cells , *STRESS granules , *LIFE sciences , *TAUOPATHIES - Abstract
The generation of retinal models from human induced pluripotent stem cells holds significant potential for advancing our understanding of retinal development, neurodegeneration, and the in vitro modeling of neurodegenerative disorders. The retina, as an accessible part of the central nervous system, offers a unique window into these processes, making it invaluable for both study and early diagnosis. This study investigates the impact of the Frontotemporal Dementia-linked IVS 10 + 16 MAPT mutation on retinal development and function using 2D and 3D retinal models derived from human induced pluripotent stem cells. Our findings reveal that the MAPT mutation leads to delayed retinal cell differentiation and maturation, with tau-mutant disease models exhibiting sustained higher expression of retinal progenitor cell markers and a reduced presence of post-mitotic neurons. Both 2D and 3D tau-mutant retinal models demonstrated an imbalance in tau isoforms, favoring 4R tau, along with increased tau phosphorylation, altered neurite morphology, and impaired cytoskeletal maturation. These changes are associated with impaired synaptic development, reduced neuronal connectivity, and enhanced cellular stress responses, including the increased formation of stress granules, markers of apoptosis and autophagy, and the presence of intracellular toxic tau aggregates. This study highlights the value of retinal models derived from human induced pluripotent stem cells in exploring the mechanisms underlying retinal pathology associated with tau mutations. These models offer essential insights into the development of therapeutic strategies for neurodegenerative diseases characterized by tau aggregation. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
31. Tau oligomers impair memory and synaptic plasticity through the cellular prion protein.
- Author
-
Balducci, Claudia, Orsini, Franca, Cerovic, Milica, Beeg, Marten, Rocutto, Beatrice, Dacomo, Letizia, Masone, Antonio, Busani, Eleonora, Raimondi, Ilaria, Lavigna, Giada, Chen, Po-Tao, Leva, Susanna, Colombo, Laura, Zucchelli, Chiara, Musco, Giovanna, Kanaan, Nicholas M., Gobbi, Marco, Chiesa, Roberto, Fioriti, Luana, and Forloni, Gianluigi
- Subjects
- *
RECOGNITION (Psychology) , *ALZHEIMER'S disease , *SURFACE plasmon resonance , *TAUOPATHIES , *MEDICAL sciences - Abstract
Deposition of abnormally phosphorylated tau aggregates is a central event leading to neuronal dysfunction and death in Alzheimer's disease (AD) and other tauopathies. Among tau aggregates, oligomers (TauOs) are considered the most toxic. AD brains show significant increase in TauOs compared to healthy controls, their concentration correlating with the severity of cognitive deficits and disease progression. In vitro and in vivo neuronal TauO exposure leads to synaptic and cognitive dysfunction, but their mechanisms of action are unclear. Evidence suggests that the cellular prion protein (PrPC) may act as a mediator of TauO neurotoxicity, as previously proposed for β-amyloid and α-synuclein oligomers. To investigate whether PrPC mediates TauO detrimental activities, we compared their effects on memory and synaptic plasticity in wild type (WT) and PrPC knockout (Prnp0/0) mice. Intracerebroventricular injection of TauOs significantly impaired recognition memory in WT but not in Prnp0/0 mice. Similarly, TauOs inhibited long-term potentiation in acute hippocampal slices from WT but not Prnp0/0 mice. Surface plasmon resonance indicated a high-affinity binding between TauOs and PrPC with a KD of 20–50 nM. Immunofluorescence analysis of naïve and PrPC-overexpressing HEK293 cells exposed to TauOs showed a PrPC dose-dependent association of TauOs with cells over time, and their co-localization with PrPC on the plasma membrane and in intracellular compartments, suggesting PrPC-may play a role in TauO internalization. These findings support the concept that PrPC mediates the detrimental activities of TauOs through a direct interaction, suggesting that targeting this interaction might be a promising therapeutic strategy for AD and other tauopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
32. Functional classification of tauopathy strains reveals the role of protofilament core residues.
- Author
-
Vaquer-Alicea, Jaime, Manon, Victor A., Bommareddy, Vaibhav, Kunach, Peter, Gupta, Ankit, Monistrol, Jim, Perez, Valerie A., Hung Tri Tran, Saez-Calveras, Nil, Siling Du, Batra, Sushobhna, Stoddard, Daniel, White III, Charles L., Joachimiak, Lukasz A., Shahmoradian, Sarah H., and Diamond, Marc I.
- Subjects
- *
ALZHEIMER'S disease , *FLUORESCENT proteins , *TAUOPATHIES , *TAU proteins , *ANIMAL experimentation - Abstract
Distinct tau amyloid assemblies underlie diverse tauopathies but defy rapid classification. Cell and animal experiments indicate tau functions as a prion, as different strains propagated in cells cause unique, transmissible neuropathology after inoculation. Strain amplification requires compatibility of the monomer and amyloid template. We used cryo-electron microscopy to study one cell-based yellow fluorescent protein (YFP)-tagged strain, resolving its amyloid nature. We then used sequential alanine (Ala) substitution (scan) within tau repeat domain (RD) to measure incorporation to preexisting tau RD-YFP aggregates. This robustly discriminated strains, defining sequences critical for monomer incorporation. We then created 3R/4R or 4R wild-type RD (amino acids 246 to 408) biosensors. Ala scan of recombinant tau seeds with the Alzheimer's disease (AD) fold matched that of AD homogenate. We scanned 22 brain lysates comprising four tauopathies. This clustered cases by neuropathological syndrome, revealed the role of amino acids in protofilament folds, and allowed strain discrimination based on amino acid requirements for prion replication. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
33. Microglial double stranded DNA accumulation induced by DNase II deficiency drives neuroinflammation and neurodegeneration.
- Author
-
Li, Ling-jie, Liang, Shi-yu, Sun, Xiao-ying, Zhu, Jie, Niu, Xiao-yun, Du, Xiao-yu, Huang, Ya-ru, and Liu, Rui-tian
- Subjects
- *
TYPE I interferons , *ALZHEIMER'S disease , *MEDICAL sciences , *TAUOPATHIES , *NEURODEGENERATION - Abstract
Background: Deoxyribonuclease 2 (DNase II) is pivotal in the clearance of cytoplasmic double stranded DNA (dsDNA). Its deficiency incurs DNA accumulation in cytoplasm, which is a hallmark of multiple neurodegenerative diseases. Our previous study showed that neuronal DNase II deficiency drove tau hyperphosphorylation and neurodegeneration (Li et al., Transl Neurodegener 13:39, 2024). Although it has been verified that DNase II participates in type I interferons (IFN-I) mediated autoinflammation and senescence in peripheral systems, the role of microglial DNase II in neuroinflammation and neurodegenerative diseases such as Alzheimer's disease (AD) is still unknown. Methods: The levels of microglial DNase II in triple transgenic AD mice (3xTg-AD) were measured by immunohistochemistry. The cognitive performance of microglial DNase II deficient WT and AD mice was determined using the Morris water maze test, Y-maze test, novel object recognition test and open filed test. To investigate the impact of microglial DNase II deficiency on microglial morphology, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway and IFN-I pathway, neuroinflammation, synapses loss, amyloid pathology and tauopathy, the levels of cGAS-STING and IFN-I pathway related protein, gliosis and proinflammatory cytokines, synaptic protein, complement protein, Aβ levels, phosphorylated tau in the brains of the microglial DNase II deficient WT and AD mice were evaluated by immunolabeling, immunoblotting, q-PCR or ELISA. Results: We found that the levels of DNase II were significantly decreased in the microglia of 3xTg-AD mice. Microglial DNase II deficiency altered microglial morphology and transcriptional signatures, activated the cGAS-STING and IFN-I pathway, initiated neuroinflammation, led to synapse loss via complement-dependent pathway, increased Aβ levels and tauopathy, and induced cognitive decline. Conclusions: Our study shows the effect of microglial DNase II deficiency and cytoplasmic accumulated dsDNA on neuroinflammation, and reveals the initiatory mechanism of AD pathology, suggesting that DNase II is a potential target for neurodegenerative diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
34. Brain-derived tau oligomer polymorphs: distinct aggregations, stability profiles, and biological activities.
- Author
-
Lo Cascio, Filippa, Park, Suhyeorn, Sengupta, Urmi, Puangmalai, Nicha, Bhatt, Nemil, Shchankin, Nikita, Jerez, Cynthia, Moreno, Naomi, Bittar, Alice, Xavier, Rhea, Zhao, Yingxin, Wang, Cankun, Fu, Hongjun, Ma, Qin, Montalbano, Mauro, and Kayed, Rakez
- Subjects
- *
LEWY body dementia , *PROGRESSIVE supranuclear palsy , *ALZHEIMER'S disease , *TAUOPATHIES , *MICROTUBULE-associated proteins , *TAU proteins - Abstract
Aggregation of microtubule-associated tau protein is a distinct hallmark of several neurodegenerative disorders such as Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). Tau oligomers are suggested to be the primary neurotoxic species that initiate aggregation and propagate prion-like structures. Furthermore, different diseases are shown to have distinct structural characteristics of aggregated tau, denoted as polymorphs. Here, we investigate the structural and functional differences of amplified brain-derived tau oligomers (aBDTOs) from AD, DLB, and PSP. Our results indicate that the aBDTOs possess different structural and morphological features that impact neuronal function, gene regulation, and ultimately disease progression. The distinct tau oligomeric polymorphs may thus contribute to the development of clinical phenotypes and shape the progression of diseases. Our results can provide insight into developing personalized therapy to target a specific neurotoxic tau polymorph. Different aggregation characteristics and stability profiles of tau oligomeric polymorphs highlight the importance of studying the tau polymorphs to shed light on the functional differences and neuropathological heterogeneity in tauopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
35. Upregulated astrocyte HDAC7 induces Alzheimer-like tau pathologies via deacetylating transcription factor-EB and inhibiting lysosome biogenesis.
- Author
-
Ye, Jinwang, Zhong, Suyue, Wan, Huali, Guo, Xing, Yao, Xuanbao, Liu, Qiong, Chen, Liming, Wang, Jian-Zhi, and Xiao, Shifeng
- Subjects
- *
ALZHEIMER'S disease , *TAUOPATHIES , *HISTONE deacetylase , *NEUROGLIA , *PROTEOLYSIS , *TAU proteins - Abstract
Background: Astrocytes, the most abundant glial cell type in the brain, will convert into the reactive state in response to proteotoxic stress such as tau accumulation, a characteristic feature of Alzheimer's disease (AD) and other tauopathies. The formation of reactive astrocytes is partially attributed to the disruption of autophagy lysosomal signaling, and inhibiting of some histone deacetylases (HDACs) has been demonstrated to reduce the molecular and functional characteristics of reactive astrocytes. However, the precise role of autophagy lysosomal signaling in astrocytes that regulates tau pathology remains unclear. Methods: We investigated the expression of class IIa HDAC7 in astrocytes from AD patients and PS19 mice. PS19 mice were treated with AAVs expressing shRNA for HDAC7 with astrocyte-specific promoter and with a selective class IIa HDAC inhibitor, TMP195, and the effects on tau pathology, gliosis, synaptic plasticity and cognition-related behavioral performance were measured. Tau uptake and degradation assays in cultured astrocytes were utilized to investigate the role of HDAC7 on astrocyte-mediated tau clearance. Immunoprecipitation, immunofluorescence, western blotting, RT-qPCR, mass spectrometric, and luciferase reporter assay were used to identify HDAC7 substrates, modification site and related signaling pathways in astrocyte-tau clearance. We generated a new antibody to clarify the role of HDAC7-mediated signaling in AD patients and PS19 mice. Results: Here, we found that the level of histone deacetylase 7 (HDAC7) was remarkably increased in the astrocytes of AD patients and P301S tau transgenic (PS19) mice. Genetic or pharmacological inhibition of HDAC7 effectively enhanced astrocytic clearance of tau with improved cognitive functions in PS19 mice. HDAC7 could modulate astrocytic uptake and lysosomal degradation of tau proteins through a transcriptional factor EB (TFEB) acetylation-dependent manner. Specifically, deacetylation of TFEB at K310 site by HDAC7 prevented TFEB nuclear translocation with reduced lysosomal biogenesis and tau clearance in astrocytes, whereas inhibiting HDAC7 restored astrocytic TFEB acetylation level at K310 with improved tau pathology and cognitive functions in PS19 mice. Conclusions: Our findings suggest that upregulation of HDAC7 induces AD-like tau pathologies via deacetylating TFEB and inhibiting lysosomal biogenesis in astrocytes, and downregulating HDAC7-TFEB signaling is promising for arresting AD and other tauopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
36. Essential tremor with tau pathology features seeds indistinguishable in conformation from Alzheimer's disease and primary age-related tauopathy.
- Author
-
Saez-Calveras, Nil, Vaquer-Alicea, Jaime, White III, Charles L., Tak, Yogesh, Cosentino, Stephanie, Faust, Phyllis L., Louis, Elan D., and Diamond, Marc I.
- Subjects
- *
PROGRESSIVE supranuclear palsy , *CHRONIC traumatic encephalopathy , *ALZHEIMER'S disease , *AMINO acid sequence , *TAUOPATHIES - Abstract
Neurodegenerative tauopathies are characterized by the deposition of distinct fibrillar tau assemblies, whose rigid core structures correlate with defined neuropathological phenotypes. Essential tremor (ET) is a progressive neurological disorder that, in some cases, is associated with cognitive impairment and tau accumulation. In this study, we explored tau assembly conformation in ET patients with tau pathology using cytometry-based tau biosensor assays. These assays quantify the tau seeding activity present in brain homogenates by detecting the conversion of intracellular tau-fluorescent protein fusions from a soluble to an aggregated state. Pathogenic tau assemblies exhibit seeding barriers, where a specific assembly structure cannot serve as a template for a native monomer if the amino acid sequences are incompatible. We recently leveraged this species barrier to define tauopathies systematically by substituting alanine (Ala) into the tau monomer and measuring its incorporation into seeded aggregates within biosensor cells. This Ala scan precisely classified the conformation of tau seeds from various tauopathies. In this study, we analyzed 18 ET patient brains with tau pathology, detecting robust tau seeding activity in 9 (50%) of the cases, predominantly localized to the temporal pole and temporal cortex. We further examined 8 of these ET cases using the Ala scan and found that the amino acid requirements for tau monomer incorporation into aggregates seeded from ET brain homogenates were identical to those of Alzheimer's disease (AD) and primary age-related tauopathy (PART), and distinct from other tauopathies, such as corticobasal degeneration (CBD), chronic traumatic encephalopathy (CTE), and progressive supranuclear palsy (PSP). These findings indicate that in a pathologically confined subset of ET cases with significant tau pathology, tau assembly cores are identical to those seen in AD and PART. This could facilitate more precise diagnosis and targeted therapies for ET patients presenting with cognitive impairment. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
37. The MIR-NAT MAPT-AS1 does not regulate Tau expression in human neurons.
- Author
-
Policarpo, Rafaela, Wolfs, Leen, Martínez-Montero, Saul, Vandermeulen, Lina, Royaux, Ines, Van Peer, Gert, Mestdagh, Pieter, De Strooper, Bart, Sierksma, Annerieke, and d'Ydewalle, Constantin
- Subjects
- *
INDUCED pluripotent stem cells , *LINCRNA , *TAU proteins , *ALZHEIMER'S disease , *TAUOPATHIES - Abstract
The MAPT gene encodes Tau protein, a member of the large family of microtubule-associated proteins. Tau forms large insoluble aggregates that are toxic to neurons in several neurological disorders, and neurofibrillary Tau tangles represent a key pathological hallmark of Alzheimer's disease (AD) and other tauopathies. Lowering Tau expression levels constitutes a potential treatment for AD but the mechanisms that regulate Tau expression at the transcriptional or translational level are not well understood. Natural antisense transcripts (NATs) are a particular class of long non-coding RNAs (lncRNAs) that can regulate expression of their overlapping protein-coding genes at the epigenetic, transcriptional, or translational level. We and others identified a long non-coding RNA associated with the MAPT gene, named MAPT antisense 1 (MAPT-AS1). We confirmed that MAPT-AS1 is expressed in neurons in human post mortem brain tissue. To study the role of MAPT-AS1 on MAPT expression regulation, we modulated the expression of this lncRNA in human neuroblastoma cell lines and in human induced pluripotent stem cell (iPSC) derived neurons. In contrast to previous reports, we observed no changes on MAPT mRNA or Tau protein levels upon modulation of MAPT-AS1 levels in these cellular models. Our data suggest that MAPT-AS1 does not regulate Tau expression levels in human neurons in vitro. Thus, MAPT-AS1 does not represent a valuable therapeutic target to lower Tau expression in patients affected by tauopathies including AD. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
38. Resting-State EEG Alpha Rhythms Are Related to CSF Tau Biomarkers in Prodromal Alzheimer's Disease.
- Author
-
Del Percio, Claudio, Lizio, Roberta, Lopez, Susanna, Noce, Giuseppe, Carpi, Matteo, Jakhar, Dharmendra, Soricelli, Andrea, Salvatore, Marco, Yener, Görsev, Güntekin, Bahar, Massa, Federico, Arnaldi, Dario, Famà, Francesco, Pardini, Matteo, Ferri, Raffaele, Carducci, Filippo, Lanuzza, Bartolo, Stocchi, Fabrizio, Vacca, Laura, and Coletti, Chiara
- Subjects
- *
ALZHEIMER'S disease , *TAU proteins , *MAGNETIC induction tomography , *MILD cognitive impairment , *TAUOPATHIES , *CEREBROSPINAL fluid examination , *ALPHA rhythm - Abstract
Patients with mild cognitive impairment due to Alzheimer's disease (ADMCI) typically show abnormally high delta (<4 Hz) and low alpha (8–12 Hz) rhythms measured from resting-state eyes-closed electroencephalographic (rsEEG) activity. Here, we hypothesized that the abnormalities in rsEEG activity may be greater in ADMCI patients than in those with MCI not due to AD (noADMCI). Furthermore, they may be associated with the diagnostic cerebrospinal fluid (CSF) amyloid–tau biomarkers in ADMCI patients. An international database provided clinical–demographic–rsEEG datasets for cognitively unimpaired older (Healthy; N = 45), ADMCI (N = 70), and noADMCI (N = 45) participants. The rsEEG rhythms spanned individual delta, theta, and alpha frequency bands. The eLORETA freeware estimated cortical rsEEG sources. Posterior rsEEG alpha source activities were reduced in the ADMCI group compared not only to the Healthy group but also to the noADMCI group (p < 0.001). Negative associations between the CSF phospho-tau and total tau levels and posterior rsEEG alpha source activities were observed in the ADMCI group (p < 0.001), whereas those with CSF amyloid beta 42 levels were marginal. These results suggest that neurophysiological brain neural oscillatory synchronization mechanisms regulating cortical arousal and vigilance through rsEEG alpha rhythms are mainly affected by brain tauopathy in ADMCI patients. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
39. Deciphering the Role of Adrenergic Receptors in Alzheimer's Disease: Paving the Way for Innovative Therapies.
- Author
-
Miliotou, Androulla N., Kotsoni, Andria, and Zacharia, Lefteris C.
- Subjects
- *
ADRENERGIC receptors , *ALZHEIMER'S disease , *ADRENERGIC beta blockers , *ADRENERGIC agonists , *TAUOPATHIES - Abstract
Neurodegenerative diseases are currently among the most devastating diseases with no effective disease-modifying drugs in the market, with Alzheimer's disease (AD) being the most prevalent. AD is a complex multifactorial neurodegenerative disorder characterized by progressive and severe cognitive impairment and memory loss. It is the most common cause of progressive memory loss (dementia) in the elderly, and to date, there is no effective treatment to cure or slow disease progression substantially. The role of adrenergic receptors in the pathogenesis of Alzheimer's disease and other tauopathies is poorly understood or investigated. Recently, some studies indicated a potential benefit of drugs acting on the adrenergic receptors for AD and dementias, although due to the heterogeneity of the drug classes used, the results on the whole remain inconclusive. The scope of this review article is to comprehensively review the literature on the possible role of adrenergic receptors in neurodegenerative diseases, stemming from the use of agonists and antagonists including antihypertensive and asthma drugs acting on the adrenergic receptors, but also from animal models and in vitro models where these receptors have been studied. Ultimately, we hope to obtain a better understanding of the role of these receptors, identify the gaps in knowledge, and explore the possibility of repurposing such drugs for AD, given their long history of use and safety. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
40. Proteostasis as a fundamental principle of Tau immunotherapy.
- Author
-
Cruz, Esteban, Nisbet, Rebecca M, Padmanabhan, Pranesh, Waardenberg, Ashley J van, Graham, Mark E, Nkajja, Godfrey, Tapaswi, Swara, Connor, Bradley J, Robinson, Phil, and Götz, Jürgen
- Subjects
- *
TAU proteins , *ALZHEIMER'S disease , *TAUOPATHIES , *MICROTUBULE-associated proteins , *NEURODEGENERATION , *POST-translational modification - Abstract
The microtubule-associated protein Tau is a driver of neuronal dysfunction in Alzheimer's disease and other tauopathies. In this process, Tau initially undergoes subtle changes to its abundance, subcellular localization and a vast array of post-translational modifications including phosphorylation that progressively result in the protein's somatodendritic accumulation and dysregulation of multiple Tau-dependent cellular processes. Given the various loss- and gain-of-functions of Tau in disease and the brain-wide changes in the proteome that characterize tauopathies, we asked whether targeting Tau would restore the alterations in proteostasis observed in disease. Therefore, by phage display, we generated a novel pan-Tau antibody, RNJ1, that preferentially binds human Tau and neutralizes proteopathic seeding activity in multiple cell lines and benchmarked it against a clinically tested pan-Tau antibody, HJ8.5 (murine version of tilavonemab). We then evaluated both antibodies, alone and in combination, in the K3 tauopathy mouse model, showing reduced Tau pathology and improvements in neuronal function following 14 weekly treatments, without obtaining synergy for the combination. These effects were more pronounced in female mice. To investigate the molecular mechanisms contributing to improvements in neuronal function, we employed quantitative proteomics, phosphoproteomics and kinase prediction analysis to first establish alterations in K3 mice relative to wild-type controls at the proteome level. In female K3 mice, we found 342 differentially abundant proteins, which are predominantly involved in metabolic and microtubule-associated processes, strengthening previously reported findings of defects in several functional domains in multiple tauopathy models. We next asked whether antibody-mediated Tau target engagement indirectly affects levels of deregulated proteins in the K3 model. Importantly, both immunotherapies, in particular RNJ1, induced abundance shifts towards a restoration to wild-type levels (proteostasis). A total of 257 of 342 (∼75%) proteins altered in K3 were closer in abundance to wild-type levels after RNJ1 treatment, and 73% after HJ8.5 treatment. However, the magnitude of these changes was less pronounced than that observed with RNJ1. Furthermore, analysis of the phosphoproteome showed an even stronger restoration effect with RNJ1, with ∼82% of altered phosphopeptides in K3 showing a shift to wild-type levels, and 75% with HJ8.5. Gene set over-representation analysis further confirmed that proteins undergoing restoration are involved in biological pathways affected in K3 mice. Together, our study suggests that a Tau immunotherapy-induced restoration of proteostasis links target engagement and treatment efficacy. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
41. Novel Role of Pin1-Cis P-Tau-ApoE Axis in the Pathogenesis of Preeclampsia and Its Connection with Dementia.
- Author
-
Amabebe, Emmanuel, Huang, Zheping, Jash, Sukanta, Krishnan, Balaji, Cheng, Shibin, Nakashima, Akitoshi, Li, Yitong, Li, Zhixong, Wang, Ruizhi, Menon, Ramkumar, Zhou, Xiao Zhen, Lu, Kun Ping, and Sharma, Surendra
- Subjects
ALZHEIMER'S disease ,TAUOPATHIES ,TAU proteins ,APOLIPOPROTEIN E ,NEURODEGENERATION - Abstract
Preeclampsia (preE) is a severe multisystem hypertensive syndrome of pregnancy associated with ischemia/hypoxia, angiogenic imbalance, apolipoprotein E (ApoE)-mediated dyslipidemia, placental insufficiency, and inflammation at the maternal–fetal interface. Our recent data further suggest that preE is associated with impaired autophagy, vascular dysfunction, and proteinopathy/tauopathy disorder, similar to neurodegenerative diseases such as Alzheimer's disease (AD), including the presence of the cis stereo-isoform of phosphorylated tau (cis P-tau), amyloid-β, and transthyretin in the placenta and circulation. This review provides an overview of the factors that may lead to the induction and accumulation of cis P-tau-like proteins by focusing on the inactivation of peptidyl-prolyl cis–trans isomerase (Pin1) that catalyzes the cis to trans isomerization of P-tau. We also highlighted the novel role of the Pin1-cis P-tau-ApoE axis in the development of preE, and propagation of cis P-tau-mediated abnormal protein aggregation (tauopathy) from the placenta to cerebral tissues later in life, leading to neurodegenerative conditions. In the case of preE, proteinopathy/tauopathy may interrupt trophoblast differentiation and induce cell death, similar to the events occurring in neurons. These events may eventually damage the endothelium and cause systemic features of disorders such as preE. Despite impressive research and therapeutic advances in both fields of preE and neurodegenerative diseases, further investigation of Pin1-cis P-tau and ApoE-related mechanistic underpinnings may unravel novel therapeutic options, and new transcriptional and proteomic markers. This review will also cover genetic polymorphisms in the ApoE alleles leading to dyslipidemia induction that may regulate the pathways causing preE or dementia-like features in the reproductive age or later in life, respectively. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
42. Modifying reaction time tasks parameters in the automated IntelliCage identifies heightened impulsivity and impaired attention in the 3xTg-AD model of Alzheimer's disease.
- Author
-
Judd, Jessica M., Winslow, Wendy, McDonough, Ian, Mistry, Faizan, and Velazquez, Ramon
- Subjects
BIOLOGICAL models ,TAU proteins ,REPEATED measures design ,TASK performance ,ALZHEIMER'S disease ,RESEARCH funding ,PHOSPHORYLATION ,T-test (Statistics) ,DATA analysis ,PREFRONTAL cortex ,ENZYME-linked immunosorbent assay ,NEUROINFLAMMATION ,DESCRIPTIVE statistics ,IMPULSIVE personality ,ATTENTION ,MICE ,TAUOPATHIES ,ANIMAL behavior ,ANIMAL experimentation ,COGNITION disorders ,THREONINE ,ONE-way analysis of variance ,STATISTICS ,REACTION time ,DATA analysis software ,AMYLOID beta-protein precursor ,SERINE ,GENOTYPES - Abstract
Background: The 3xTg-AD transgenic mouse model of Alzheimer's disease (AD) is an important tool to investigate the relationship between development of pathological amyloid-β (Aβ) and tau, neuroinflammation, and cognitive impairments. Traditional behavioral tasks assessing aspects of learning and memory, such as mazes requiring spatial navigation, unfortunately suffer from several shortcomings, including the stress of human handling and not probing species-typical behavior. The automated IntelliCage system was developed to circumvent such issues by testing mice in a social environment while measuring multiple aspects of cognition. Water consumption can serve as a primary motivator for task engagement. Once animals adapt to the cage and can access water, mice can be subjected to operant tasks. Each of the four corners of a cage contains doors to manipulate access to water, visual LED cues, and a valve allowing administration of an air puff. Previously, we detected significant impairments in 3xTg-AD mice in the IntelliCage, however a high failure rate and genotypical differences in water motivation were observed. Methods: Here, we implemented an IntelliCage paradigm where mice underwent progressively more difficult reaction time tasks to assess attention and impulsivity, behaviors mediated by the prefrontal cortex. Mice were placed in the IntelliCage at 11.5 months of age, which corresponds with the presence of widespread pathology. Results: As the difficulty of the reaction time tasks increased, 3xTg-AD mice exhibited lower percent Correct Responses than NonTg. When implementing varying pre-cue durations, where animals are required to wait between the initiation of the trial and the LED turning on (which then requires a nose-poke to access water), 3xTg-AD mice prematurely nose-poked on trials requiring a longer delay before a second nose poke would allow water access, demonstrating heightened impulsivity. The presence of soluble and insoluble fractions of cortical Aβ40 and 42, and phosphorylated tau epitopes threonine 181 and serine 396 confirmed the presence of neuropathological hallmarks in 3xTg-AD mice. Conclusion: Together, this study describes a novel protocol that overcomes motivational differences and detects attention and impulsivity deficits in 3xTg-AD mice utilizing the IntelliCage. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
43. Mitochondrial bioenergetics stimulates autophagy for pathological MAPT/Tau clearance in tauopathy neurons.
- Author
-
Jia, Nuo, Ganesan, Dhasarathan, Guan, Hongyuan, Jeong, Yu Young, Han, Sinsuk, Rajapaksha, Gavesh, Nissenbaum, Marialaina, Kusnecov, Alexander W., and Cai, Qian
- Subjects
FLUORESCENCE resonance energy transfer ,CYTOCHROME oxidase ,DORSAL root ganglia ,TAU proteins ,TAUOPATHIES - Abstract
Hyperphosphorylation and aggregation of MAPT (microtubule-associated protein tau) is a pathogenic hallmark of tauopathies and a defining feature of Alzheimer disease (AD). Pathological MAPT/tau is targeted by macroautophagy/autophagy for clearance after being sequestered within autophagosomes, but autophagy dysfunction is indicated in tauopathy. While mitochondrial bioenergetic deficits have been shown to precede MAPT/tau pathology in tauopathy brains, it is unclear whether energy metabolism deficiency is involved in the pathogenesis of autophagy defects. Here, we reveal that stimulation of anaplerotic metabolism restores defective oxidative phosphorylation (OXPHOS) in tauopathy neurons which, strikingly, leads to pronounced MAPT/tau clearance by boosting autophagy functionality through enhancements of mitochondrial biosynthesis and supply of phosphatidylethanolamine for autophagosome biogenesis. Furthermore, early anaplerotic stimulation of OXPHOS elevates autophagy activity and attenuates MAPT/tau pathology, thereby counteracting memory impairment in tauopathy mice. Taken together, our study sheds light on a pivotal role of mitochondrial bioenergetic deficiency in tauopathy-related autophagy defects and suggests a new therapeutic strategy to prevent the buildup of pathological MAPT/tau in AD and other tauopathy diseases. Abbreviation: AA: antimycin A; AD, Alzheimer disease; ATP, adenosine triphosphate; AV, autophagosome/autophagic vacuole; AZ, active zone; Baf-A1: bafilomycin A
1 ; CHX, cycloheximide; COX, cytochrome c oxidase; DIV, days in vitro; DRG, dorsal root ganglion; ETN, ethanolamine; FRET, Förster/fluorescence resonance energy transfer; FTD, frontotemporal dementia; Gln, glutamine; HA: hydroxylamine; HsMAPT/Tau, human MAPT; IMM, inner mitochondrial membrane; LAMP1, lysosomal-associated membrane protein 1; LIs, lysosomal inhibitors; MDAV, mitochondria-derived autophagic vacuole; MmMAPT/Tau, murine MAPT; NFT, neurofibrillary tangle; OCR, oxygen consumption rate; Omy: oligomycin; OXPHOS, oxidative phosphorylation; PPARGC1A/PGC-1alpha: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PE, phosphatidylethanolamine; phospho-MAPT/tau, hyperphosphorylated MAPT; PS, phosphatidylserine; PISD, phosphatidylserine decarboxylase;SQSTM1/p62, sequestosome 1; STX1, syntaxin 1; SYP, synaptophysin; Tg, transgenic; TCA, tricarboxylic acid; TEM, transmission electron microscopy. [ABSTRACT FROM AUTHOR]- Published
- 2025
- Full Text
- View/download PDF
44. Distinct regulation of Tau Monomer and aggregate uptake and intracellular accumulation in human neurons.
- Author
-
Marvian, Amir T., Strauss, Tabea, Tang, Qilin, Tuck, Benjamin J., Keeling, Sophie, Rüdiger, Daniel, Mirzazadeh Dizaji, Negar, Mohammad-Beigi, Hossein, Nuscher, Brigitte, Chakraborty, Pijush, Sutherland, Duncan S., McEwan, William A., Köglsperger, Thomas, Zahler, Stefan, Zweckstetter, Markus, Lichtenthaler, Stefan F., Wurst, Wolfgang, Schwarz, Sigrid, and Höglinger, Günter
- Subjects
- *
HEPARAN sulfate proteoglycans , *BIOLOGICAL transport , *BINDING site assay , *LIFE sciences , *TAUOPATHIES - Abstract
Background: The prion-like spreading of Tau pathology is the leading cause of disease progression in various tauopathies. A critical step in propagating pathologic Tau in the brain is the transport from the extracellular environment and accumulation inside naïve neurons. Current research indicates that human neurons internalize both the physiological extracellular Tau (eTau) monomers and the pathological eTau aggregates. However, similarities or differences in neuronal transport mechanisms between Tau species remain elusive. Method: Monomers, oligomers, and fibrils of recombinant 2N4R Tau were produced and characterized by biochemical and biophysical methods. A neuronal eTau uptake and accumulation assay was developed for human induced pluripotent stem cell-derived neurons (iPSCNs) and Lund human mesencephalic cells (LUHMES)-derived neurons. Mechanisms of uptake and cellular accumulation of eTau species were studied by using small molecule inhibitors of endocytic mechanisms and siRNAs targeting Tau uptake mediators. Results: Extracellular Tau aggregates accumulated more than monomers in human neurons, mainly due to the higher efficiency of small fibrillar and soluble oligomeric aggregates in intraneuronal accumulation. A competition assay revealed a distinction in the neuronal accumulation between physiological eTau Monomers and pathology-relevant aggregates, suggesting differential transport mechanisms. Blocking heparan sulfate proteoglycans (HSPGs) with heparin only inhibited the accumulation of eTau aggregates, whereas monomers' uptake remained unaltered. At the molecular level, the downregulation of genes involved in HSPG synthesis exclusively blocked neuronal accumulation of eTau aggregates but not monomers, suggesting its role in the transport of pathologic Tau. Moreover, the knockdown of LRP1, as a receptor of Tau, mainly reduced the accumulation of monomeric form, confirming its involvement in Tau's physiological transport. Conclusion: These data propose that despite the similarity in the cellular mechanism, the uptake and accumulation of eTau Monomers and aggregates in human neurons are regulated by different molecular mediators. Thus, they address the possibility of targeting the pathological spreading of Tau aggregates without disturbing the probable physiological or non-pathogenic transport of Tau Monomers. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. Paradoxical attenuation of early amyloid-induced cognitive impairment and synaptic plasticity in an aged APP/Tau bigenic rat model.
- Author
-
Emmerson, Joshua T., Do Carmo, Sonia, Lavagna, Agustina, Huang, Chunwei, Wong, Tak Pan, Martinez-Trujillo, Julio C., and Cuello, A. Claudio
- Subjects
- *
AMYLOID beta-protein , *LABORATORY rats , *ALZHEIMER'S disease , *TAUOPATHIES , *NEUROPLASTICITY - Abstract
The combination of amyloid beta and tau pathologies leads to tau-mediated neurodegeneration in Alzheimer's disease. However, the relative contributions of amyloid beta and tau peptide accumulation to the manifestation of the pathological phenotype in the early stages, before the overt deposition of plaques and tangles, are still unclear. We investigated the longitudinal pathological effects of combining human-like amyloidosis and tauopathy in a novel transgenic rat model, coded McGill-R-APPxhTau. We compared the effects of individual and combined amyloidosis and tauopathy in transgenic rats by assessing the spatiotemporal progression of Alzheimer's-like amyloid and tau pathologies using biochemical and immunohistochemical methods. Extensive behavioral testing for learning and memory was also conducted to evaluate cognitive decline. Additionally, we investigated brain inflammation, neuronal cell loss, as well as synaptic plasticity through acute brain slice electrophysiological recordings and Western blotting. Evaluation of Alzheimer's-like amyloidosis and tauopathy, at the initial stages, unexpectedly revealed that the combination of amyloid pathology with the initial increment in phosphorylated tau exerted a paradoxical corrective effect on amyloid-induced cognitive impairments and led to a compensatory-like restoration of synaptic plasticity as revealed by electrophysiological evidence, compared to monogenic transgenic rats with amyloidosis or tauopathy. We discovered elevated CREB phosphorylation and increased expression of postsynaptic proteins as a tentative explanation for the improved hippocampal synaptic plasticity. However, this tau-induced protective effect on synaptic function was transient. As anticipated, at more advanced stages, the APPxhTau bigenic rats exhibited aggravated tau and amyloid pathologies, cognitive decline, increased neuroinflammation, and tau-driven neuronal loss compared to monogenic rat models of Alzheimer's-like amyloid and tau pathologies. The present findings propose that the early accumulation of phosphorylated tau may have a transient protective impact on the evolving amyloid pathology-derived synaptic impairments. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Dementia with lewy bodies patients with high tau levels display unique proteome profiles.
- Author
-
Greally, Sinead, Kumar, Mukesh, Schlaffner, Christoph, van der Heijden, Hanne, Lawton, Elisabeth S., Biswas, Deeptarup, Berretta, Sabina, Steen, Hanno, and Steen, Judith A.
- Subjects
- *
LEWY body dementia , *ALZHEIMER'S disease , *POST-translational modification , *TAUOPATHIES , *LIFE sciences - Abstract
Background: Clinical studies have long observed that neurodegenerative disorders display a range of symptoms and pathological features and, in some cases, overlap, suggesting that these diseases exist on a spectrum. Dementia with Lewy Bodies (DLB), a synucleinopathy, is a prominent example, where symptomatic similarities with tauopathy, Alzheimer's disease, are observed. Although tau pathology has been observed in DLB, the interplay between tau and α-synuclein is poorly understood at a molecular level. Methods: Quantitative mass spectrometry analysis was used to measure protein abundance in the insoluble fraction from cortical brain tissue from pathologically diagnosed DLB subjects (n = 30) and age-matched controls (n = 29). Using tau abundance, we stratified the DLB subjects into two subgroups termed DLBTau+ (higher abundance) and DLBTau− (lower abundance). We conducted proteomic analysis to characterize and compare the cortical proteome of DLB subjects exhibiting elevated tau, as well as the molecular modifications of tau and α-synuclein to explore the dynamic between tau and α-synuclein pathology in these patients. Results: Proteomic analyses revealed distinct global protein dysregulations in DLBTau+ and DLBTau− subjects when compared to controls. Notably, DLBTau+ patients exhibited increased levels of tau, along with ubiquitin, and APOE, indicative of cortical proteome alterations associated with elevated tau. Comparing DLBTau+ and DLBTau− groups, we observed significant upregulation of cytokine signaling and metabolic pathways in DLBTau− patients, while DLBTau+ subjects showed increases in protein ubiquitination processes and regulation of vesicle-mediated transport. Additionally, we examined the post-translational modification patterns of tau and α-synuclein. Our analysis revealed distinct phosphorylation and ubiquitination sites on α-synuclein between groups. Moreover, we observed increased modifications on tau specifically within the DLBTau+ subgroup. Conclusion: This molecular-level data supports the idea of neurodegenerative disease as a continuum of diseases with distinct PTM profiles DLBTau+ and DLBTau− patients in comparison to AD. These findings further emphasize the importance of identifying specific and tailored therapeutic approaches targeting the involved proteopathies in the neurodegenerative disease spectrum. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. Drug Discovery and Screening Tool Development for Tauopathies by Focusing on Pathogenic Tau Repeat 3 Oligomers.
- Author
-
Yoon, Soljee, Kim, Hye Yun, Park, Sohui, Cha, Minhae, Kim, Kyeonghwan, Lee, Songmin, Kim, JiMin, Bhang, Saeyun, and Kim, YoungSoo
- Subjects
- *
ALZHEIMER'S disease , *TAUOPATHIES , *DRUG discovery , *MEDICAL screening , *HIGH throughput screening (Drug development) , *TAU proteins - Abstract
Comprehending early amyloidogenesis is essential for the development of effective therapeutic strategies. In tauopathies like Alzheimer's disease (AD), the abnormal accumulation of tau protein is initiated by pathological tau seeds. Mounting evidence implies that the microtubule binding domain, consisting of three to four repeats, plays a pivotal role in this process, yet the exact region driving the formation of pathogenic species needs to be further scrutinized. Here, we chemically synthesized individual tau repeats to identify those exhibiting pathogenic prion‐like characteristics. Notably, repeat 3 (R3) displayed a remarkable propensity to polymerize, form toxic filaments, and induce cognitive impairment, even in the absence of an aggregation‐promoting inducer, highlighting its physiological relevance. Additionally, oligomeric R3 was identified as a particularly pathological form, prompting the establishment of a screening platform. Through screening, tolcapone was found to possess therapeutic efficacy against pathological tau aggregates in PS19 transgenic mice. This screening platform provides a valuable avenue for identifying compounds that selectively interact with peptides implicated in the progression of tauopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Amyloid-β oligomers increase the binding and internalization of tau oligomers in human synapses.
- Author
-
Kadamangudi, Shrinath, Marcatti, Michela, Zhang, Wen-Ru, Fracassi, Anna, Kayed, Rakez, Limon, Agenor, and Taglialatela, Giulio
- Subjects
- *
ALZHEIMER'S disease , *LIFE sciences , *TEMPORAL lobe , *TAUOPATHIES , *MEMBRANE proteins , *TAU proteins - Abstract
In Alzheimer's disease (AD), the propagation and spreading of CNS tau pathology closely correlates with cognitive decline, positioning tau as an attractive therapeutic target. Amyloid beta (Aβ) has been strongly implicated in driving tau spread, whereas primary tauopathies such as primary age-related tauopathy (PART)—which lack Aβ pathology—exhibit limited tau spread and minimal-to-no cognitive decline. Emerging evidence converges on a trans-synaptic mechanism of tau spread, facilitated by the transfer of misfolded tau aggregates (e.g. soluble oligomers). However, it is unclear whether Aβ oligomers modulate the binding and internalization of tau oligomers in human synapses. Our translationally focused paradigms utilize post-mortem brain specimens from Control, PART, and AD patients. Synaptosomes isolated from the temporal cortex of all three groups were incubated with preformed recombinant tauO (rtauO), ± preformed recombinant AβO (rAβO), and oligomer binding/internalization was quantified via flow cytometry following proteinase K (PK) digestion of surface-bound oligomers. TauO-synapse interactions were visualized using EM immunogold. Brain-derived tau oligomers (BDTO) from AD and PART PBS-soluble hippocampal fractions were co-immunoprecipitated and analyzed via mass spectrometry to compare synaptic tauO interactomes in primary and secondary tauopathies, thereby inferring the role of Aβ. AD synaptosomes, enriched in endogenous Aβ pathology, exhibited increased rtauO internalization compared to PART synaptosomes. This observation was mirrored in Control synaptosomes, where recombinant rAβO significantly increased rtauO binding and internalization. PK pre-treatment abolished this effect, implicating synaptic membrane proteins in AβO-mediated tauO internalization. While both PART and AD BDTO were broadly enriched in synaptic proteins, AD BDTO exhibited differential enrichment of endocytic proteins across pre- and post-synaptic compartments, whereas PART BDTO showed no significant synaptic enrichment. This study demonstrates that Aβ oligomers enhance tau oligomer binding and drive its internalization through synaptic membrane proteins. These findings offer novel mechanistic insights underlying pathological tau spreading directly within human synapses and emphasize the therapeutic potential of targeting Aβ-tau interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. Tau seeds catalyze fibril-type structures from GFP tau biosensor cells.
- Author
-
Wang, Jinliang, Williams, Christopher K., DeTure, Michael A., Magaki, Shino D., Dickson, Dennis W., Vinters, Harry V., and Seidler, Paul M.
- Subjects
- *
ALZHEIMER'S disease , *TAUOPATHIES , *FLUORESCENT proteins , *CHIMERIC proteins , *DISEASE progression , *TAU proteins - Abstract
Fibril-type aggregates of tau occur in Alzheimer's disease (AD) and dozens of tauopathies. Fibrils catalyze aggregation by prion-like seeding, which in part underlies disease progression. Seeding by recombinant and brain-derived tau fibrils is measured using biosensor cells that express aggregation-prone tau mutants fused with fluorescent reporter proteins. Seeding results in a punctated phenotype that is well established, but evidence that fluorescent tau fusion proteins from biosensor cells assemble into fibril-type structures is lacking. We investigated the effects of seeding on fibril formation by biosensor cells. Fluorescent punctated cell phenotypes that were catalyzed persisted with varying stabilities. Seeded cells bearing punctated phenotypes yielded sarkosyl-insoluble fibrils, although non-seeded cells did not. ImmunoEM of cell-purified fibrils shows that GFP localizes to the proteolytically sensitive fuzzy coat of tau fibrils. The presented data offer compelling evidence that fluorescent puncta are fibril-type aggregates of tau that result from prion-like seeding. [Display omitted] • Cells expressing tau fused with the GFP reporter undergo aggregation through seeding • Fluorescent tau aggregates are amyloid-like fibrils • GFP reporter localizes to tau fuzzy coat rendering it proteolytically sensitive Biosensor cells are commonly used in protein aggregation studies. Wang et al. demonstrated that biosensor cells expressing tau undergo fibril formation, confirming the amyloid-like nature of tau seeding. These findings set the stage for investigating seeding fidelity and cellular factors that contribute to fibril polymorphisms. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. dGAE(297–391) Tau Fragment Promotes Formation of Chronic Traumatic Encephalopathy‐Like Tau Filaments.
- Author
-
Kitoka, Kristine, Lends, Alons, Kucinskas, Gytis, Bula, Anna Lina, Krasauskas, Lukas, Smirnovas, Vytautas, Zilkova, Monika, Kovacech, Branislav, Skrabana, Rostislav, Hritz, Jozef, and Jaudzems, Kristaps
- Subjects
- *
CHRONIC traumatic encephalopathy , *TAU proteins , *CELL aggregation , *TAUOPATHIES , *NUCLEAR magnetic resonance spectroscopy - Abstract
The microtubule‐associated protein tau forms disease‐specific filamentous aggregates in several different neurodegenerative diseases. In order to understand how tau undergoes misfolding into a specific filament type and to control this process for drug development purposes, it is crucial to study in vitro tau aggregation methods and investigate the structures of the obtained filaments at the atomic level. Here, we used the tau fragment dGAE, which aggregates spontaneously, to seed the formation of full‐length tau filaments. The structures of dGAE and full‐length tau filaments were investigated by magic‐angle spinning (MAS) solid‐state NMR, showing that dGAE allows propagation of a chronic traumatic encephalopathy (CTE)‐like fold to the full‐length tau. The obtained filaments efficiently seeded tau aggregation in HEK293T cells. This work demonstrates that in vitro preparation of disease‐specific types of full‐length tau filaments is feasible. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.