1
|
Wang S, Qi C, Rajpurohit C, Ghosh B, Xiong W, Wang B, Qi Y, Hwang SH, Hammock BD, Li H, Gan L, Zheng H. Inhibition of soluble epoxide hydrolase confers neuroprotection and restores microglial homeostasis in a tauopathy mouse model. Mol Neurodegener 2025; 20:44. [PMID: 40264187 PMCID: PMC12016400 DOI: 10.1186/s13024-025-00844-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND The epoxyeicosatrienoic acids (EETs) are derivatives of the arachidonic acid metabolism with anti-inflammatory activities. However, their efficacy is limited due to the rapid hydrolysis by soluble epoxide hydrolase (sEH). Inhibition of sEH has been shown to stabilize the EETs and reduce neuroinflammation in Aβ mouse models of Alzheimer's disease (AD). However, the role of the sEH-EET signaling pathway in other CNS cell types and neurodegenerative conditions are less understood. METHODS Here we investigated the mechanisms and functional role of the sEH-EET axis in tauopathy by treating PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice. This was followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical analysis, and behavioral assessments. Additionally, we examined the effects of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons exhibiting seeding-induced Tau inclusions. RESULTS sEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglial reactivity. snRNA-seq revealed that TPPU treatment upregulated genes involved in actin cytoskeleton and excitatory synaptic pathways. Treatment of human iPSC-derived neurons with TPPU enhanced synaptic density without affecting Tau accumulation, suggesting a cell-autonomous neuroprotective effect of sEH blockade. Furthermore, sEH inhibition reversed disease-associated and interferon-responsive microglial states in PS19 mice, while EET supplementation promoted Tau phagocytosis and clearance in primary microglia cultures. CONCLUSION These findings demonstrate that sEH blockade or EET augmentation confers therapeutic benefit in neurodegenerative tauopathies by simultaneously targeting neuronal and microglial pathways.
Collapse
Affiliation(s)
- Shuo Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chuangye Qi
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chetan Rajpurohit
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Baijayanti Ghosh
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
di Lorenzo D, Bisi N, Bucci R, Ennen I, Lo Presti L, Dodero V, Brandt R, Ongeri S, Gelmi ML, Tonali N. Application of modular isoxazoline-β 2,2-amino acid-based peptidomimetics as chemical model systems for studying the tau misfolding. iScience 2025; 28:112272. [PMID: 40264794 PMCID: PMC12013406 DOI: 10.1016/j.isci.2025.112272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/29/2024] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
Tau is a microtubule-associated protein essential for regulating microtubule dynamics and axonal transport in neurons. In tauopathies, the transition of tau from a physiological to a pathological form remains unclear, though the hexapeptides PHF6 and PHF6∗ are key in triggering aggregation. These sequences are shielded by a β-hairpin structure in the native state but expose hydrophobic residues during misfolding, promoting self-assembly. This study employs a non-natural β2-amino acid to induce PHF6 and PHF6∗ into either extended or β-hairpin conformations. The extended form triggers tau aggregation without additives, acting as a seed-competent monomer model system. Conversely, the β-hairpin preserves tau in a soluble monomeric state. Additionally, a β-hairpin mimic inspired by Hsp90 showed potential as a chaperone mimic and inhibitor of tau aggregation, offering insights into corrective folding and aggregation modulation in neuronal environments.
Collapse
Affiliation(s)
- Davide di Lorenzo
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400 Orsay, France
- Dipartimento di Scienze Farmaceutiche, DISFARM, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Nicolo Bisi
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400 Orsay, France
- Department of Neurobiology, Osnabrück University, Barbarastrasse 11, D-49076 Osnabrück, Germany
| | - Raffaella Bucci
- Dipartimento di Scienze Farmaceutiche, DISFARM, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Inga Ennen
- Department of Physics, Bielefeld University Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Leonardo Lo Presti
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Veronica Dodero
- Department of Chemistry, Physical and Biophysical Chemistry, Bielefeld University Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Roland Brandt
- Department of Neurobiology, Osnabrück University, Barbarastrasse 11, D-49076 Osnabrück, Germany
| | - Sandrine Ongeri
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400 Orsay, France
| | - Maria-Luisa Gelmi
- Dipartimento di Scienze Farmaceutiche, DISFARM, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Nicolo Tonali
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17 av. des Sciences, 91400 Orsay, France
- CEA Saclay, DRF/JOLIOT/DMTS/SIMoS/LPEM, 91191 Gif-sur-Yvette, France
| |
Collapse
|
3
|
Li W, Dasgupta A, Yang K, Wang S, Hemandhar-Kumar N, Chepyala SR, Yarbro JM, Hu Z, Salovska B, Fornasiero EF, Peng J, Liu Y. Turnover atlas of proteome and phosphoproteome across mouse tissues and brain regions. Cell 2025; 188:2267-2287.e21. [PMID: 40118046 DOI: 10.1016/j.cell.2025.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/27/2025] [Accepted: 02/21/2025] [Indexed: 03/23/2025]
Abstract
Understanding how proteins in different mammalian tissues are regulated is central to biology. Protein abundance, turnover, and post-translational modifications such as phosphorylation are key factors that determine tissue-specific proteome properties. However, these properties are challenging to study across tissues and remain poorly understood. Here, we present Turnover-PPT, a comprehensive resource mapping the abundance and lifetime of 11,000 proteins and 40,000 phosphosites in eight mouse tissues and various brain regions using advanced proteomics and stable isotope labeling. We reveal tissue-specific short- and long-lived proteins, strong correlations between interacting protein lifetimes, and distinct impacts of phosphorylation on protein turnover. Notably, we discover a remarkable pattern of turnover changes for peroxisome proteins in specific tissues and that phosphorylation regulates the stability of neurodegeneration-related proteins, such as Tau and α-synuclein. Thus, Turnover-PPT provides fundamental insights into protein stability, tissue dynamic proteotypes, and functional protein phosphorylation and is accessible via an interactive web-based portal at https://yslproteomics.shinyapps.io/tissuePPT.
Collapse
Affiliation(s)
- Wenxue Li
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Abhijit Dasgupta
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ka Yang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shisheng Wang
- Department of General Surgery and Liver Transplant Center, Proteomics-Metabolomics Analysis Platform, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Nisha Hemandhar-Kumar
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Surendhar R Chepyala
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jay M Yarbro
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhenyi Hu
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Barbora Salovska
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Eugenio F Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA; Department of Biomedical Informatics & Data Science, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
4
|
Mirabelli M, Chiefari E, Arcidiacono B, Salatino A, Pascarella A, Morelli M, Credendino SC, Brunetti FS, Di Vito A, Greco A, Huin V, Nicoletti F, Pierantoni GM, Fedele M, Aguglia U, Foti DP, Brunetti A. HMGA1 deficiency: a pathogenic link between tau pathology and insulin resistance. EBioMedicine 2025; 115:105700. [PMID: 40233659 DOI: 10.1016/j.ebiom.2025.105700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Growing evidence links tau-related neurodegeneration with insulin resistance and type 2 diabetes (T2D), though the underlying mechanisms remain unclear. Our previous research identified HMGA1 as crucial for insulin receptor (INSR) expression, with defects in the HMGA1 gene associated with insulin resistance and T2D. Here, we explore HMGA1 deficiency as a potential contributor to tauopathies, such as Alzheimer's disease (AD), and its connection to insulin resistance. METHODS Immunoblot analyses, protein-DNA interaction studies, ChIP-qPCR, and reporter gene assays were conducted in human and mouse neuronal cell models. Tau immunohistochemistry, behavioural studies, and brain glucose metabolism were analysed in Hmga1-knockout mice. Additionally, a case-control study investigated the relationship between HMGA1 and tau pathology in patients with tauopathy, carrying or not the HMGA1 rs146052672 variant, known to reduce HMGA1 protein levels and increase the risk of insulin resistance and T2D. FINDINGS We show that HMGA1 regulates tau protein expression primarily through the specific repression of MAPT gene transcription. In both human neuronal cells and primary mouse neurons, tau mRNA and protein levels were inversely correlated with HMGA1 expression. This inverse relationship was further confirmed in the brain of Hmga1-knockout mice, where tau was overexpressed, INSR was downregulated, and brain glucose uptake was impaired. Additionally, the rs146052672 variant was more common in patients with tauopathy (12/69, 17.4%) than in controls (10/200, 5.0%) (p = 0.001), and carriers of this variant exhibited more severe disease progression and poorer therapeutic outcomes. INTERPRETATION These findings suggest that HMGA1 deficiency may drive tau pathology, linking tauopathies to insulin resistance and providing new insights into the relationship between metabolic and neurodegenerative disorders. Furthermore, our observation that over 17% of individuals with tauopathy exhibit a deficit in HMGA1 protein production could have significant clinical implications, potentially guiding the development of therapeutic strategies targeting this specific defect. FUNDING See acknowledgements section.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Alessandro Salatino
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Angelo Pascarella
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Maurizio Morelli
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Sara C Credendino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Francesco S Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Anna Di Vito
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Adelaide Greco
- Interdepartmental Centre of Veterinary Radiology, University of Naples "Federico II", Naples, Italy
| | - Vincent Huin
- University of Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, UMR-S1172, Team Alzheimer & Tauopathies, F-59000, Lille, France
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University of Rome "Sapienza", Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Giovanna M Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology, CNR, Naples, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Daniela P Foti
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| | - Antonio Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| |
Collapse
|
5
|
Bisi N, Pinzi L, Rastelli G. Selective imaging probes for differential detection of pathological tau polymorphs in tauopathies. Drug Discov Today 2025; 30:104352. [PMID: 40216294 DOI: 10.1016/j.drudis.2025.104352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/12/2025] [Accepted: 04/04/2025] [Indexed: 04/20/2025]
Abstract
Tauopathies, including Alzheimer's disease (AD), Pick's disease (PiD), progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD), are characterized by the misfolding and pathological aggregation of the tau protein, leading to neurodegeneration. Although the pathogenesis of these diseases is still a matter for debate, the formation of amyloid inclusions still represents the only histopathological hallmark available. Tau inclusions are not the same in terms of structure and morphology, and different tauopathies are characterized by different polymorphs. Remarkably, the selective detection of these polymorphs is crucial for differential diagnosis, disease monitoring and evaluation of the potential harmfulness of polymorphs, with a significant impact on drug discovery. This review discusses recent advances in the development of imaging probes designed for the selective detection of pathological tau forms associated with specific tauopathies. We explore the application of compounds that can target tau polymorphs characteristic of AD, PiD, PSP and CBD. In particular, we focus on discussing the probes' selectivity and sensitivity in distinguishing between the different tauopathy-associated polymorphs in preclinical settings. The progress and the weaknesses in this field are discussed, to guide the researchers in identifying accurate and potent probes for the selective diagnosis of these different neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicolò Bisi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy.
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
| |
Collapse
|
6
|
Sugiyama M, Kosik KS, Panagiotou E. Geometry based prediction of tau protein sites and motifs associated with misfolding and aggregation. Sci Rep 2025; 15:10283. [PMID: 40133414 PMCID: PMC11937417 DOI: 10.1038/s41598-025-93304-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Recent studies of tau proteins point to specific sites or motifs along the protein related to its misfolding and aggregation propensity, which is associated with neurodegenerative diseases of structure-dependent pathology. In this manuscript we employ topology and geometry to analyze the local structure of tau proteins obtained from the Protein Data Bank. Our results show that mathematical topology/geometry of cryo-EM structures alone identify the PGGG motifs, and the PHF6(*) motifs as sites of interest and reveal a geometrical hierarchy of the PGGG motifs that differs for 3R+4R and 4R tauopathies. By employing the Local Topological Free Energy (LTE), we find that progressive supranuclear palsy (PSP) and globular glial tauopathy (GGT) have the highest LTE values around residues 302-305, which are inside the jR2R3 peptide and in the vicinity of the 301 site, experimentally associated with aggregation. By extending the LTE definition to estimate a global topological free energy, we find that the jR2R3 peptide of PSP and GGT, has in fact the lowest global topological free energy among other tauopathies. These results point to a possible correlation between the global topological free energy of parts of the protein and the LTE of specific sites.
Collapse
Affiliation(s)
- Masumi Sugiyama
- Department of Mathematics, University of Tennessee at Chattanooga, Chattanooga, TN, 37403, USA
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM2), Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Kenneth S Kosik
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Eleni Panagiotou
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, 85281, USA.
| |
Collapse
|
7
|
Chakra Bortty J, Chakraborty GS, Noman IR, Batra S, Das J, Bishnu KK, Tarafder MTR, Islam A. A Novel Diagnostic Framework with an Optimized Ensemble of Vision Transformers and Convolutional Neural Networks for Enhanced Alzheimer's Disease Detection in Medical Imaging. Diagnostics (Basel) 2025; 15:789. [PMID: 40150131 PMCID: PMC11941083 DOI: 10.3390/diagnostics15060789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Alzheimer's disease (AD) is a progressive, neurodegenerative disorder, which causes memory loss and loss of cognitive functioning, along with behavioral changes. Early detection is important to delay disease progression, timely intervention and to increase patients' and caregivers' quality of life (QoL). One of the major and primary challenges for preventing any disease is to identify the disease at the initial stage through a quick and reliable detection process. Different researchers across the world are still working relentlessly, coming up with significant solutions. Artificial intelligence-based solutions are putting great importance on identifying the disease efficiently, where deep learning with medical imaging is highly being utilized to develop disease detection frameworks. In this work, a novel and optimized detection framework has been proposed that comes with remarkable performance that can classify the level of Alzheimer's accurately and efficiently. Methods: A powerful vision transformer model (ViT-B16) with three efficient Convolutional Neural Network (CNN) models (VGG19, ResNet152V2, and EfficientNetV2B3) has been trained with a benchmark dataset, 'OASIS', that comes with a high volume of brain Magnetic Resonance Images (MRI). Results: A weighted average ensemble technique with a Grasshopper optimization algorithm has been designed and utilized to ensure maximum performance with high accuracy of 97.31%, precision of 97.32, recall of 97.35, and F1 score of 0.97. Conclusions: The work has been compared with other existing state-of-the-art techniques, where it comes with high efficiency, sensitivity, and reliability. The framework can be utilized in IoMT infrastructure where one can access smart and remote diagnosis services.
Collapse
Affiliation(s)
- Joy Chakra Bortty
- Department of Computer Science, Westcliff University, 17877 Von Karman Ave 4th Floor, Irvine, CA 92614, USA; (J.C.B.); (A.I.)
| | - Gouri Shankar Chakraborty
- Department of Computer Science and Engineering, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Inshad Rahman Noman
- Department of Computer Science, California State University, 5151 State University Dr, Los Angeles, CA 90032, USA; (I.R.N.); (K.K.B.)
| | - Salil Batra
- Department of Computer Science and Engineering, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Joy Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Kanchon Kumar Bishnu
- Department of Computer Science, California State University, 5151 State University Dr, Los Angeles, CA 90032, USA; (I.R.N.); (K.K.B.)
| | | | - Araf Islam
- Department of Computer Science, Westcliff University, 17877 Von Karman Ave 4th Floor, Irvine, CA 92614, USA; (J.C.B.); (A.I.)
| |
Collapse
|
8
|
Mahendran TS, Singh A, Srinivasan S, Jennings CM, Neureuter C, Gindra BH, Parekh SH, Banerjee PR. Decoupling Phase Separation and Fibrillization Preserves Activity of Biomolecular Condensates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643977. [PMID: 40166274 PMCID: PMC11957012 DOI: 10.1101/2025.03.18.643977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Age-dependent transition of metastable, liquid-like protein condensates to amyloid fibrils is an emergent phenomenon of numerous neurodegeneration-linked protein systems. A key question is whether the thermodynamic forces underlying reversible phase separation and maturation to irreversible amyloids are distinct and separable. Here, we address this question using an engineered version of the microtubule-associated protein Tau, which forms biochemically active condensates. Liquid-like Tau condensates exhibit rapid aging to amyloid fibrils under quiescent, cofactor-free conditions. Tau condensate interface promotes fibril nucleation, impairing their activity to recruit tubulin and catalyze microtubule assembly. Remarkably, a small molecule metabolite, L-arginine, selectively impedes condensate-to-fibril transition without perturbing phase separation in a valence and chemistry-specific manner. By heightening the fibril nucleation barrier, L-arginine counteracts age-dependent decline in the biochemical activity of Tau condensates. These results provide a proof-of-principle demonstration that small molecule metabolites can enhance the metastability of protein condensates against a liquid-to-amyloid transition, thereby preserving condensate function.
Collapse
Affiliation(s)
- Tharun Selvam Mahendran
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Anurag Singh
- Department of Physics, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Sukanya Srinivasan
- Department of Physics, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Christian M. Jennings
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Christian Neureuter
- Department of Physics, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Bhargavi H. Gindra
- Department of Physics, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Sapun H. Parekh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Priya R. Banerjee
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
- Department of Physics, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
| |
Collapse
|
9
|
Parhizkar S, Holtzman DM. The night's watch: Exploring how sleep protects against neurodegeneration. Neuron 2025; 113:817-837. [PMID: 40054454 PMCID: PMC11925672 DOI: 10.1016/j.neuron.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Sleep loss is often regarded as an early manifestation of neurodegenerative diseases given its common occurrence and link to cognitive dysfunction. However, the precise mechanisms by which sleep disturbances contribute to neurodegeneration are not fully understood, nor is it clear why some individuals are more susceptible to these effects than others. This review addresses critical unanswered questions in the field, including whether sleep disturbances precede or result from neurodegenerative diseases, the functional significance of sleep changes during the preclinical disease phase, and the potential role of sleep homeostasis as an adaptive mechanism enhancing resilience against cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Samira Parhizkar
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
10
|
Uliassi E, Bolognesi ML, Milelli A. Targeting Tau Protein with Proximity Inducing Modulators: A New Frontier to Combat Tauopathies. ACS Pharmacol Transl Sci 2025; 8:654-672. [PMID: 40109749 PMCID: PMC11915046 DOI: 10.1021/acsptsci.4c00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 03/22/2025]
Abstract
Dysregulation of correct protein tau homeostasis represents the seed for the development of several devastating central nervous system disorders, known as tauopathies, that affect millions of people worldwide. Despite massive public and private support to research funding, these diseases still represent unmet medical needs. In fact, the tau-targeting tools developed to date have failed to translate into the clinic. Recently, taking advantage of the modes that nature uses to mediate the flow of information in cells, researchers have developed a new class of molecules, called proximity-inducing modulators, which exploit spatial proximity to modulate protein function(s) and redirect cellular processes. In this perspective, after a brief discussion about tau protein and the classic tau-targeting approaches, we will discuss the different classes of proximity-inducing modulators developed so far and highlight the applications to modulate tau protein's function and tau-induced toxicity.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Andrea Milelli
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d'Augusto 237, Rimini 47921, Italy
| |
Collapse
|
11
|
Tangavelou K, Jiang S, Dadras S, Hulse JP, Sanchez K, Bondu V, Villaseñor Z, Mandell M, Peabody J, Chackerian B, Bhaskar K. Pathological tau activates inflammatory nuclear factor-kappa B (NF-κB) and pT181-Qβ vaccine attenuates NF-κB in PS19 tauopathy mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642500. [PMID: 40161741 PMCID: PMC11952447 DOI: 10.1101/2025.03.10.642500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Tau regulates neuronal integrity. In tauopathy, phosphorylated tau detaches from microtubules and aggregates, and is released into the extracellular space. Microglia are the first responders to the extracellular tau, a danger/damage-associated molecular pattern (DAMP), which can be cleared by proteostasis and activate innate immune response gene expression by nuclear factor-kappa B (NF-κB). However, longitudinal NF-κB activation in tauopathies and whether pathological tau (pTau) contributes to NF-κB activity is unknown. Here, we tau oligomers from human Alzheimer's disease brain (AD-TO) activate NF-κB in mouse microglia and macrophages reducing the IκBα via promoting its secretion in the extracellular space. NF-κB activity peaks at 9- and 11-months age in PS19Luc + and hTauLuc + mice, respectively. Reducing pTau via pharmacological (DOX), genetic ( Mapt -/- ) or antibody-mediated neutralization (immunization with pT181-Qβ vaccine) reduces NF-κB activity, and together suggest pTau is a driver of NF-κB and chronic neuroinflammation tauopathies. Summary Neuronal tau activates microglial NF-κB constitutively by secreting its inhibitor IκBα. NF-κB activation in PS19Luc + and hTauLuc + mice peaks at 9- and 11-months of age, respectively. Neutralizing pTau with pT181-Qβ vaccine (targeting phosphorylated threonine 181 tau) alleviates NF-κB activity in tauopathy mice.
Collapse
|
12
|
Giannakis A, Sioka C, Kloufetou E, Konitsiotis S. Cognitive impairment in Parkinson's disease and other parkinsonian syndromes. J Neural Transm (Vienna) 2025; 132:341-355. [PMID: 39614911 DOI: 10.1007/s00702-024-02865-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 03/03/2025]
Abstract
In this narrative review, we address mild cognitive impairment, a frequent complication of Parkinson's disease (PD) and atypical parkinsonian disorders (APDs). Recent diagnostic criteria have blurred the lines between PD and dementia with Lewy bodies (DLB), particularly in the cognitive domain. Additionally, atypical parkinsonian syndromes like progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) often present with significant cognitive decline. Even multiple system atrophy (MSA) can be associated with cognitive impairment in some cases. Several biomarkers, including imaging techniques, such brain magnetic resonance imaging (MRI) and fluorodeoxyglucose positron emission tomography (FDG-PET), as well as pathological proteins either of the cerebrospinal fluid (CSF), such as Tau, amyloid beta, and synuclein, or of the serum, such as neurofilament light chain (Nfl) are more and more often utilized in the early differential diagnosis of APDs. The complex interplay between these conditions and the evolving understanding of their underlying pathologies highlight the need for further research to refine diagnostic criteria, possibly incorporate the new findings from the biomarker's field into the diagnostic criteria and develop targeted therapeutic strategies.
Collapse
Affiliation(s)
- Alexandros Giannakis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Stavrou Niarchou Av., University Campus, Ioannina, Greece.
| | - Chrissa Sioka
- Department of Nuclear Medicine, Faculty of Medicine, University of Ioannina, Stavrou Niarchou Av., University Campus, Ioannina, Greece
| | - Eugenia Kloufetou
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Stavrou Niarchou Av., University Campus, Ioannina, Greece
| | - Spiridon Konitsiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Stavrou Niarchou Av., University Campus, Ioannina, Greece
| |
Collapse
|
13
|
Awasthi S, Tiwari PC, Awasthi S, Dwivedi A, Srivastava S. Mechanistic role of proteins and peptides in Management of Neurodegenerative Disorders. Neuropeptides 2025; 110:102505. [PMID: 39965449 DOI: 10.1016/j.npep.2025.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Proteins and peptides have emerged as significant contributors in the management of neurodegenerative disorders due to their diverse biological functions. These biomolecules influence various cellular processes, including cellular repair, inflammation reduction, and neuronal survival, which are crucial for mitigating the effects of diseases such as Alzheimer's, Parkinson's, and Amyotrophic Lateral Sclerosis (ALS). By interacting with specific cellular receptors, proteins and peptides like neurotrophic factors, cytokines, and enzyme inhibitors promote neurogenesis, reduce oxidative stress, and enhance synaptic plasticity. Nevertheless, till certain limitations and challenges do exist to deliver these fragile therapeutic bioactives. Moreover, targeted delivery systems, such as nanoparticles and biomolecular carriers, are being developed to improve the bioavailability and specificity of these protein-based therapeutics, ensuring efficient crossing of the blood-brain barrier. This review explores the mechanistic pathways through which these biomolecules act, emphasizing their potential to modify disease progression and improve the quality of life in patients with neurodegenerative conditions. Overall, proteins and peptides are not only seen as promising therapeutic agents but also as foundational tools in advancing personalized medicine in the field of neurodegenerative disorders.
Collapse
Affiliation(s)
- Saumya Awasthi
- Institute of Pharmacy, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Barabanki, Uttar Pradesh 225003, India
| | | | - Srishti Awasthi
- Institute of Pharmaceutical Sciences and Research, Unnao 209859, India
| | - Arpit Dwivedi
- Institute of Pharmaceutical Sciences and Research, Unnao 209859, India
| | - Shikha Srivastava
- Institute of Pharmacy, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Barabanki, Uttar Pradesh 225003, India.
| |
Collapse
|
14
|
Kim BS, Hwang I, Ko HR, Kim YK, Kim HJ, Seo SW, Choi Y, Lim S, Kim YK, Nie S, Ye K, Park JC, Lee Y, Jo DG, Lee SE, Kim D, Cho SW, Ahn JY. EBP1 potentiates amyloid β pathology by regulating γ-secretase. NATURE AGING 2025; 5:486-503. [PMID: 39779912 DOI: 10.1038/s43587-024-00790-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
The abnormal deposition of amyloid β (Aβ), produced by proteolytic cleavage events of amyloid precursor protein involving the protease γ-secretase and subsequent polymerization into amyloid plaques, plays a key role in the neuropathology of Alzheimer's disease (AD). Here we show that ErbB3 binding protein 1 (EBP1)/proliferation-associated 2G4 (PA2G4) interacts with presenilin, a catalytic subunit of γ-secretase, inhibiting Aβ production. Mice lacking forebrain Ebp1/Pa2g4 recapitulate the representative phenotypes of late-onset sporadic AD, displaying an age-dependent increase in Aβ deposition, amyloid plaques and cognitive dysfunction. In postmortem brains of patients with AD and 5x-FAD mice, we found that EBP1 is proteolytically cleaved by asparagine endopeptidase at N84 and N204 residues, compromising its inhibitory effect on γ-secretase, increasing Aβ aggregation and neurodegeneration. Accordingly, injection of AAV2-Ebp1 wild-type or an asparagine endopeptidase-uncleavable mutant into the brains of 5x-FAD mice decreased Aβ generation and alleviated the behavioral impairments. Thus, our study suggests that EBP1 acts as an inhibitor of γ-secretase on amyloid precursor protein cleavage and preservation of functional EBP1 could be a therapeutic strategy for AD.
Collapse
Affiliation(s)
- Byeong-Seong Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Inwoo Hwang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Hyo Rim Ko
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Young Kwan Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yujung Choi
- Center for Brain Disorders, Brain Science Institute Korea Institute of Science and Technology (KIST), Seoul, Korea
- Department of Life Sciences, Korea University, Seoul, Korea
| | - Sungsu Lim
- Center for Brain Disorders, Brain Science Institute Korea Institute of Science and Technology (KIST), Seoul, Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Yun Kyung Kim
- Center for Brain Disorders, Brain Science Institute Korea Institute of Science and Technology (KIST), Seoul, Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Shuke Nie
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jong-Chan Park
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Yunjong Lee
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Seung Eun Lee
- Research Animal Resources Center, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Daesik Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea.
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
15
|
Cawley NX, Zhou R, Farhat NM, Iben J, Alexander DM, Luke RA, Padilla CJ, Mohamed HO, Albert OK, Robbins KP, Rahhal S, Do AD, Berry‐Kravis E, Cologna SM, Liu F, Porter FD. Elevated Cerebrospinal Fluid Total Tau in Niemann-Pick Disease Type C1: Correlation With Clinical Severity and Response to Therapeutic Interventions. J Inherit Metab Dis 2025; 48:e70016. [PMID: 40064165 PMCID: PMC11893204 DOI: 10.1002/jimd.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
Niemann-Pick disease, type C1 (NPC1) is an inborn error of intracellular cholesterol transport. Impaired function of NPC1 leads to endolysosomal accumulation of unesterified cholesterol, which results in progressive neurodegeneration. Although the age of onset is variable, classical NPC1 is a pediatric disease. Identification of biomarkers that correlate with clinical phenotype and respond to therapeutic interventions will be essential for developing effective therapeutic interventions. Aβ peptides and Tau protein are primary components of amyloid plaques and neurofibrillary tangles, respectively, which are major pathological features in neurodegenerative disorders. Cerebrospinal fluid (CSF) levels of total Tau, a biomarker of axonal damage, were elevated ~3-fold (p < 0.0001) in 106 individuals with Niemann-Pick disease, type C1, relative to age-appropriate comparison samples. Baseline CSF total Tau levels correlated with clinical measures of disease severity. Specifically, CSF total Tau levels decreased with increased age of neurological onset (rs = -0.42, FDR adj. p < 0.0001) and increased with increased Annual Severity Increment Score (rs = 0.52, FDR adj. p < 0.0001). Baseline CSF total Tau levels were decreased 40% (p = 0.0066) in individuals being treated with miglustat, and longitudinal analysis substantiated this observation with a 40% decrease (p < 0.0001, 95% CI 32%-47.4%). Longitudinal analysis also showed a significant (p = 0.004) decrease of 19% (95% CI 7%-30%) in total Tau levels associated with intrathecal 2-hydroxypropyl-β-cyclodextrin therapy. These data show that CSF total Tau levels are significantly increased in individuals with NPC1, positively correlated with increased disease severity, and respond to therapeutic interventions.
Collapse
Affiliation(s)
- Niamh X. Cawley
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Ruyu Zhou
- Applied and Computational Mathematics and StatisticsUniversity of Notre DameIndianaUSA
| | - Nicole M. Farhat
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - James Iben
- Molecular Genomics CoreEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Derek M. Alexander
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Rachel A. Luke
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Cameron J. Padilla
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Hibaaq O. Mohamed
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Orsolya K. Albert
- Departments of Pediatrics, Neurological Sciences, and Anatomy and Cell BiologyRush University Medical CenterChicagoIllinoisUSA
| | - Kendall P. Robbins
- Departments of Pediatrics, Neurological Sciences, and Anatomy and Cell BiologyRush University Medical CenterChicagoIllinoisUSA
| | - Samar Rahhal
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - An Dang Do
- Unit on Cellular Stress in Development and Diseases, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| | - Elizabeth Berry‐Kravis
- Departments of Pediatrics, Neurological Sciences, and Anatomy and Cell BiologyRush University Medical CenterChicagoIllinoisUSA
| | - Stephanie M. Cologna
- Department of Chemistry and Laboratory of Integrative NeuroscienceUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Fang Liu
- Applied and Computational Mathematics and StatisticsUniversity of Notre DameIndianaUSA
| | - Forbes D. Porter
- Section on Molecular Dysmorphology, Division of Translational MedicineEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
16
|
Lazaros K, Adam S, Krokidis MG, Exarchos T, Vlamos P, Vrahatis AG. Non-Invasive Biomarkers in the Era of Big Data and Machine Learning. SENSORS (BASEL, SWITZERLAND) 2025; 25:1396. [PMID: 40096210 PMCID: PMC11902325 DOI: 10.3390/s25051396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/19/2025]
Abstract
Invasive diagnostic techniques, while offering critical insights into disease pathophysiology, are often limited by high costs, procedural risks, and patient discomfort. Non-invasive biomarkers represent a transformative alternative, providing diagnostic precision through accessible biological samples or physiological data, including blood, saliva, breath, and wearable health metrics. They encompass molecular and imaging approaches, revealing genetic, epigenetic, and metabolic alterations associated with disease states. Furthermore, advances in breathomics and gut microbiome profiling further expand their diagnostic scope. Even with their strengths in terms of safety, cost-effectiveness, and accessibility, non-invasive biomarkers face challenges in achieving monitoring sensitivity and specificity comparable to traditional clinical approaches. Computational advancements, particularly in artificial intelligence and machine learning, are addressing these limitations by uncovering complex patterns in multi-modal datasets, enhancing diagnostic accuracy and facilitating personalized medicine. The present review integrates recent innovations, examines their clinical applications, highlights their limitations and provides a concise overview of the evolving role of non-invasive biomarkers in precision diagnostics, positioning them as a compelling choice for large-scale healthcare applications.
Collapse
Affiliation(s)
| | | | - Marios G. Krokidis
- Bioinformatics and Human Electrophysiology Laboratory, Department of Informatics, Ionian University, 49100 Corfu, Greece
| | | | | | | |
Collapse
|
17
|
Wang S, Qi C, Rajpurohit C, Ghosh B, Xiong W, Wang B, Qi Y, Hwang SH, Hammock BD, Li H, Gan L, Zheng H. Inhibition of Soluble Epoxide Hydrolase Confers Neuroprotection and Restores Microglial Homeostasis in a Tauopathy Mouse Model. RESEARCH SQUARE 2025:rs.3.rs-6038641. [PMID: 40060041 PMCID: PMC11888548 DOI: 10.21203/rs.3.rs-6038641/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Background The epoxyeicosatrienoic acids (EETs) are derivatives of the arachidonic acid metabolism with anti-inflammatory activities. However, their efficacy is limited due to the rapid hydrolasis by the soluble epoxide hydrolase (sEH). Accordingly, inhibition of sEH has been shown to stabilize the EETs and dampen neuroinflammation in Aβ mouse models of Alzheimer's disease (AD). However, the role of the sEH-EET signaling pathway in other cell types of the CNS and in other neurodegenerative conditions are less understood. Methods Here we examined the mechanisms and the functional role of the sEH-EET axis in tauopathy by treating the PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice, followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical characterization, and behavioral analysis. We also tested the effect of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons that develop seeding-induced Tau inclusions. Results We show that sEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglia reactivity. snRNA-seq revealed that TPPU treatment resulted in the upregulation of actin cytoskeleton and excitatory synaptic pathway genes. Treating the human iPSC-derived neurons with TPPU led to enhanced synaptic density without affecting Tau accumulation, indicating a cell-autonomous effect of sEH blockade in neuroprotection. Further, sEH inhibition reversed disease-associated and interferon-response microglia states in PS19 mice and EET supplementation enhanced Tau phagocytosis and clearance in primary microglia cultures. Conclusion These findings demonstrate that sEH blockade or EET augmentation confer therapeutic benefit against neurodegenerative tauopathies through parallel targeting of neuronal and microglial pathways.
Collapse
Affiliation(s)
- Shuo Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Chuangye Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | | | - Baijayanti Ghosh
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
18
|
Reddi Sree R, Kalyan M, Anand N, Mani S, Gorantla VR, Sakharkar MK, Song BJ, Chidambaram SB. Newer Therapeutic Approaches in Treating Alzheimer's Disease: A Comprehensive Review. ACS OMEGA 2025; 10:5148-5171. [PMID: 39989768 PMCID: PMC11840625 DOI: 10.1021/acsomega.4c05527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 02/25/2025]
Abstract
Alzheimer's disease (AD) is an aging-related irreversible neurodegenerative disease affecting mostly the elderly population. The main pathological features of AD are the extracellular Aβ plaques generated by APP cleavage through the amyloidogenic pathway, the intracellular neurofibrillary tangles (NFT) resulting from the hyperphosphorylated tau proteins, and cholinergic neurodegeneration. However, the actual causes of AD are unknown, but several studies suggest hereditary mutations in PSEN1 and -2, APOE4, APP, and the TAU genes are the major perpetrators. In order to understand the etiology and pathogenesis of AD, various hypotheses are proposed. These include the following hypotheses: amyloid accumulation, tauopathy, inflammation, oxidative stress, mitochondrial dysfunction, glutamate/excitotoxicity, cholinergic deficiency, and gut dysbiosis. Currently approved therapeutic interventions are donepezil, galantamine, and rivastigmine, which are cholinesterase inhibitors (ChEIs), and memantine, which is an N-methyl-d-aspartate (NMDA) antagonist. These treatment strategies focus on only symptomatic management of AD by attenuating symptoms but not regeneration of neurons or clearance of Aβ plaques and hyperphosphorylated Tau. This review focuses on the pathophysiology, novel therapeutic targets, and disease-altering treatments such as α-secretase modulators, active immunotherapy, passive immunotherapy, natural antioxidant products, nanomaterials, antiamyloid therapy, tau aggregation inhibitors, transplantation of fecal microbiota or stem cells, and microtubule stabilizers that are in clinical trials or still under investigation.
Collapse
Affiliation(s)
- Radhakrishna Reddi Sree
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Manjunath Kalyan
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre
for Experimental Pharmacology & Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department
of Pharmacology, American University of
Antigua College of Medicine, University Park, Jabberwock Beach Road, Coolidge, Antigua, Barbuda
| | - Sangeetha Mani
- Department
of Pharmacognosy, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and
Research, Porur, Chennai 600116, India
| | - Vasavi Rakesh Gorantla
- Department
of Anatomical Sciences, St. George’s University School of Medicine, St. George’s University, Saint George, Grenada
| | - Meena Kishore Sakharkar
- College
of
Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Byoung-Joon Song
- Section
of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry
and Biophysics, National Institute on Alcohol
Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20892, United States
| | - Saravana Babu Chidambaram
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre
for Experimental Pharmacology & Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| |
Collapse
|
19
|
Pragati, Sarkar S. Targeted downregulation of insulin signaling restricts human tau pathogenesis by reinstating the aberrant heterochromatin loss and mTOR/4EBP/S6K pathway in Drosophila. Brain Res 2025; 1849:149347. [PMID: 39579954 DOI: 10.1016/j.brainres.2024.149347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the accumulation of paired helical filaments (PHFs)/or neurofibrillary tangles (NFTs) in neuronal/glial cells. Besides hyperphosphorylation of tau protein, aberrant heterochromatin loss and translation dysfunction have emerged as other important aspects contributing to the disease pathogenesis. We have recently reported that tissue-specific downregulation of insulin signaling or its growth-promoting downstream sub-branch effectively reinstates the tau-mediated overactivated insulin pathway, and restricts pathogenic tau hyperphosphorylation and aggregate formation. We next investigated if the downregulation of the insulin pathway or its growth-promoting downstream sub-branch makes any impact on tau-mediated aberrant heterochromatin loss and translation dysfunction. For the first time, we demonstrate that tissue-specific downregulation of insulin signaling or its growth-promoting branch effectively restricts the pathogenic tau-induced heterochromatin loss. We further report that expression of human tau in Drosophila causes induction of the mTOR/4EBP/S6K pathway and energy disbalance which gets effectively balanced upon downregulation of insulin signaling. Our findings establish an imperative role of insulin signaling in effectively mitigating various aspects of tau etiology in Drosophila ranging from hyperphosphorylation, chromatin relaxation, and translational upsurge. Our findings could be beneficial in establishing novel therapeutic options against tauopathies.
Collapse
Affiliation(s)
- Pragati
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110 021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110 021, India.
| |
Collapse
|
20
|
Chinnathambi S, Rangappa N, Chandrashekar M. Internalization of extracellular Tau oligomers in Alzheimer's disease. Adv Clin Chem 2025; 126:1-29. [PMID: 40185532 DOI: 10.1016/bs.acc.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
A key factor in the progression of Alzheimer's disease (AD) is internalization of extracellular Tau oligomers (ecTauOs) by neuroglial cells. Aberrant hyperphosphorylation of Tau results in their dissociation from microtubules and formation of toxic intracellular Tau oligomers (icTauOs). These are subsequently released to the extracellular space following neuronal dysfunction and death. Although receptor mediated internalization of these ecTauOs by other neurons, microglia and astrocytes can facilitate elimination, incomplete degradation thereof promotes inflammation, exacerbates pathologic spread and accelerates neurodegeneration. Targeting Tau oligomer degradation pathways, blocking internalization receptors, and mitigating neuroinflammation are proposed as therapeutic strategies to control Tau propagation and toxicity. This review highlights the urgent need for innovative approaches to prevent the spread of Tau pathology, emphasizing its implications for AD and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India.
| | - Nagaraj Rangappa
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Madhura Chandrashekar
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| |
Collapse
|
21
|
Uytterhoeven V, Verstreken P, Nachman E. Synaptic sabotage: How Tau and α-Synuclein undermine synaptic health. J Cell Biol 2025; 224:e202409104. [PMID: 39718548 DOI: 10.1083/jcb.202409104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/07/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
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.
Collapse
Affiliation(s)
- Valerie Uytterhoeven
- Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research , Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Patrik Verstreken
- Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research , Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Eliana Nachman
- Vlaams Instituut voor Biotechnologie Center for Brain and Disease Research , Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Watamura N, Foiani MS, Bez S, Bourdenx M, Santambrogio A, Frodsham C, Camporesi E, Brinkmalm G, Zetterberg H, Patel S, Kamano N, Takahashi M, Rueda-Carrasco J, Katsouri L, Fowler S, Turkes E, Hashimoto S, Sasaguri H, Saito T, Islam AS, Benner S, Endo T, Kobayashi K, Ishida C, Vendruscolo M, Yamada M, Duff KE, Saido TC. In vivo hyperphosphorylation of tau is associated with synaptic loss and behavioral abnormalities in the absence of tau seeds. Nat Neurosci 2025; 28:293-307. [PMID: 39719507 PMCID: PMC11802456 DOI: 10.1038/s41593-024-01829-7] [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: 05/28/2023] [Accepted: 10/23/2024] [Indexed: 12/26/2024]
Abstract
Tau pathology is a hallmark of several neurodegenerative diseases, including frontotemporal dementia and Alzheimer's disease. However, the sequence of events and the form of tau that confers toxicity are still unclear, due in large part to the lack of physiological models of tauopathy initiation and progression in which to test hypotheses. We have developed a series of targeted mice expressing frontotemporal-dementia-causing mutations in the humanized MAPT gene to investigate the earliest stages of tauopathy. MAPTInt10+3G>A and MAPTS305N;Int10+3G>A lines show abundant hyperphosphorylated tau in the hippocampus and entorhinal cortex, but they do not develop seed-competent fibrillar structures. Accumulation of hyperphosphorylated tau was accompanied by neurite degeneration, loss of viable synapses and indicators of behavioral abnormalities. Our results demonstrate that neuronal toxicity can occur in the absence of fibrillar, higher-order structures and that tau hyperphosphorylation is probably involved in the earliest etiological events in tauopathies showing isoform ratio imbalance.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan.
- UK Dementia Research Institute at University College London, London, UK.
| | - Martha S Foiani
- UK Dementia Research Institute at University College London, London, UK.
| | - Sumi Bez
- UK Dementia Research Institute at University College London, London, UK
| | - Mathieu Bourdenx
- UK Dementia Research Institute at University College London, London, UK
| | - Alessia Santambrogio
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Claire Frodsham
- UK Dementia Research Institute at University College London, London, UK
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, London, UK
- Queen Square Institute of Neurology, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Saisha Patel
- UK Dementia Research Institute at University College London, London, UK
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | | | - Loukia Katsouri
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Stephanie Fowler
- UK Dementia Research Institute at University College London, London, UK
- Nuffield Department of Medicine, Oxford-GSK Institute of Molecular and Computational Medicine, Centre for Human Genetics, Oxford, UK
| | - Emir Turkes
- UK Dementia Research Institute at University College London, London, UK
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Pioneering Research Division, Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Dementia Pathophysiology Collaboration Unit, RIKEN Center for Brain Science, Wako, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Afm Saiful Islam
- Queen Square Institute of Neurology, University College London, London, UK
| | - Seico Benner
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | | | - Katsuji Kobayashi
- Department of Psychiatry, Awazu Neuropsychiatric Hospital, Ishikawa, Japan
| | - Chiho Ishida
- Department of Neurology, NHO Iou National Hospital, Iwade-machi, Japan
| | - Michele Vendruscolo
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Masahito Yamada
- Department of Internal Medicine, Division of Neurology, Kudanzaka Hospital, Tokyo, Japan
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
- Kanazawa University, Kanazawa, Japan
| | - Karen E Duff
- UK Dementia Research Institute at University College London, London, UK.
- Queen Square Institute of Neurology, University College London, London, UK.
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan.
| |
Collapse
|
23
|
Silva-Llanes I, Madruga E, Martínez A, Lastres-Becker I. RIPK1 expression and inhibition in tauopathies: implications for neuroinflammation and neuroprotection. Front Neurosci 2025; 18:1530809. [PMID: 39931431 PMCID: PMC11808139 DOI: 10.3389/fnins.2024.1530809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 12/11/2024] [Indexed: 02/13/2025] Open
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the alteration/aggregation of TAU protein. One of the main challenges of these diseases is that they have neither biomarkers nor pharmacological targets to stop the neurodegenerative process. Apart from the neurodegenerative process, tauopathies are also characterized by a chronic low-grade neuroinflammation process, where the receptor-interacting protein kinase 1 (RIPK1) protein plays an essential role. Our research aimed to explore the role of RIPK1 in various tauopathies. We examined mouse models of frontotemporal dementia (FTD), as well as brain tissue samples from patients with progressive supranuclear palsy (PSP), a primary form of 4R tauopathy, and Alzheimer's disease (AD), which is considered a secondary tauopathy. Our findings show elevated levels of RIPK1 mRNA levels across various forms of tauopathies, in both mouse models and human tissue samples associated with primary and secondary TAU-related disorders. Furthermore, we investigated the potential of using a RIPK1 inhibitor, known as GSK2982772, in a mouse model as a novel treatment strategy for FTD. The data showed that GSK2982772 treatment effectively reduced the reactive astrocyte response triggered by TAUP301L overexpression. However, this RIPK1 inhibitor failed to protect against the neurodegeneration caused by elevated TAUP301L levels in the hippocampal region. These results suggest that although inhibiting RIPK1 activity may help reduce TAU-related astrogliosis in the brain, the complexity of the inflammatory pathways involved could explain the absence of neuroprotective effects against TAU-induced neurodegeneration.
Collapse
Affiliation(s)
- Ignacio Silva-Llanes
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
- Instituto de Investigaciones Biomédicas “Sols-Morreale” UAM-CSIC, Madrid, Spain
| | - Enrique Madruga
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Isabel Lastres-Becker
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
- Instituto de Investigaciones Biomédicas “Sols-Morreale” UAM-CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
24
|
Renganathan A, Minaya MA, Broder M, Alfradique-Dunham I, Moritz M, Bhagat R, Marsh J, Verbeck A, Galasso G, Starr E, Agard DA, Cruchaga C, Karch CM. A novel lncRNA FAM151B-DT regulates autophagy and degradation of aggregation prone proteins. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.22.25320997. [PMID: 39974060 PMCID: PMC11838976 DOI: 10.1101/2025.01.22.25320997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neurodegenerative diseases share common features of protein aggregation along with other pleiotropic traits, including shifts in transcriptional patterns, neuroinflammation, disruptions in synaptic signaling, mitochondrial dysfunction, oxidative stress, and impaired clearance mechanisms like autophagy. However, key regulators of these pleotropic traits have yet to be identified. Here, we discovered a novel long non-coding RNA (lncRNA), FAM151B-DT, that is reduced in a stem cell model of frontotemporal dementia with tau inclusions (FTLD-tau) and in brains from FTLD-tau, progressive supranuclear palsy, Alzheimer's disease, and Parkinson's disease patients. We show that silencing FAM151B-DT in vitro is sufficient to enhance tau aggregation. To begin to understand the mechanism by which FAM151B-DT mediates tau aggregation and contributes to several neurodegenerative diseases, we deeply characterized this novel lncRNA and found that FAM151B-DT resides in the cytoplasm where it interacts with tau, α-synuclein, HSC70, and other proteins enriched in protein homeostasis. When silenced, FAM151B-DT blocks autophagy, leading to the accumulation of tau and α-synuclein. Importantly, we discovered that increasing FAM151B-DT expression is sufficient to promote autophagic flux, reduce phospho-tau and α-synuclein, and reduce tau aggregation. Overall, these findings pave the way for further exploration of FAM151B-DT as a promising molecular target for several neurodegenerative diseases.
Collapse
Affiliation(s)
- Arun Renganathan
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Miguel A. Minaya
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Matthew Broder
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | | | - Michelle Moritz
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Reshma Bhagat
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Jacob Marsh
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Anthony Verbeck
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Grant Galasso
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Emma Starr
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - David A. Agard
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
- Chan Zuckerberg Imaging Institute, Redwood City, CA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, MO
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, MO
| |
Collapse
|
25
|
Wongso H, Harada R, Furumoto S. Current Progress and Future Directions in Non-Alzheimer's Disease Tau PET Tracers. ACS Chem Neurosci 2025; 16:111-127. [PMID: 39762194 DOI: 10.1021/acschemneuro.4c00319] [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] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) and non-AD tauopathies are dominant public health issues driven by several factors, especially in the aging population. The discovery of first-generation radiotracers, including [18F]FDDNP, [11C]PBB3, [18F]flortaucipir, and the [18F]THK series, for the in vivo detection of tauopathies has marked a significant breakthrough in the fields of neuroscience and radiopharmaceuticals, creating a robust new category of labeled compounds: tau positron emission tomography (PET) tracers. Subsequently, other tau PET tracers with improved binding properties have been developed using various chemical scaffolds to target the three-repeat/four-repeat (3R/4R) tau folds in AD. In 2020, [18F]flortaucipir was approved by the U.S. Food and Drug Administration for PET imaging of tau pathology in adult patients with cognitive deficits undergoing evaluation for AD. Despite remarkable progress in the development of AD tau PET tracers, imaging agents for rare non-AD tauopathies (4R tauopathies [predominantly expressing a 4R tau isoform], involved in progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, and globular glial tauopathy, and 3R tauopathies [predominantly expressing a 3R tau isoform], such as Pick's disease) remain substantially underdeveloped. In this review, we discuss recent progress in tau PET tracer development, with particular emphasis on clinically validated tracers for AD and their potential use for non-AD tauopathies. Additionally, we highlight the critical need for further development of tau PET tracers specifically designed for non-AD tauopathies, an area that remains significantly underexplored despite its importance in advancing the understanding and diagnosis of these disorders.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Ryuichi Harada
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Shozo Furumoto
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
26
|
Fan S, Jia C, Liang M, Ren H, Zhang T, Li Q, Huang Z, Yen TC, OuYang C, Cui R, Guan H. Patterns of Tau pathology in patients with anti-IgLON5 disease visualized by Florzolotau (18F) PET. J Neurol 2025; 272:115. [PMID: 39812840 DOI: 10.1007/s00415-024-12874-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Anti-IgLON5 disease is a rare autoimmune neurological disorder with prominent Tau protein deposits in the brainstem and hypothalamus. The aim of this study was to visualize the in vivo distribution patterns of Tau protein in patients with anti-IgLON5 disease using the second-generation Tau PET tracer, Florzolotau (18F) PET imaging. METHODS Patients diagnosed with anti-IgLON5 disease were enrolled consecutively. Age- and sex-matched healthy controls (HCs) were also enrolled. The uptake of Florzolotau (18F) and 18F-FDG was assessed using both visual and semi-quantitative analysis techniques. RESULTS A total of 10 patients with anti-IgLON5 disease and 40 HCs were included in the study. All ten patients with anti-IgLON5 disease underwent Florzolotau (18F) PET scans, and five of them underwent 18F-FDG PET scans. Twenty HCs underwent Florzolotau (18F) PET scans, and the remaining 20 HCs underwent 18F-FDG PET scans. In patients with anti-IgLON5 disease, significant uptake of Florzolotau (18F) was observed predominantly in the midbrain, pons, cerebellum, caudate, and putamen. This uptake pattern was notably absent in the control group. Moreover, semi-quantitative analysis techniques demonstrated widespread hypometabolism in the cerebral cortex in patients with anti-IgLON5 disease. CONCLUSIONS This study indicates distinct Tau protein deposition patterns in patients with anti-IgLON5 disease, potentially serving as imaging biomarkers.
Collapse
Affiliation(s)
- Siyuan Fan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chenhao Jia
- Department of Nuclear Medicine, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Menglin Liang
- Department of Nuclear Medicine, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Haitao Ren
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tianhao Zhang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Qijun Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Zhaoxia Huang
- Department of Nuclear Medicine, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | | | - Chujun OuYang
- School of Computer Science, Xiangtan University, Xiangtan, Hunan, China
| | - Ruixue Cui
- Department of Nuclear Medicine, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China.
| | - Hongzhi Guan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
27
|
Rivas-Grajales AM, Han SC, Wang R, Greenstein P, Shih LC. L266V MAPT Gene Mutation Associated With Frontotemporal Dementia, Progressive Supranuclear Palsy, and Corticobasal Syndrome. J Neuropsychiatry Clin Neurosci 2025:appineuropsych20240128. [PMID: 39789940 DOI: 10.1176/appi.neuropsych.20240128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Affiliation(s)
- Ana Maria Rivas-Grajales
- Departments of Psychiatry (Rivas-Grajales) and Neurology (Han, Wang), Boston University Chobanian & Avedisian School of Medicine, Boston; Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston (Greenstein, Shih)
| | - Steve C Han
- Departments of Psychiatry (Rivas-Grajales) and Neurology (Han, Wang), Boston University Chobanian & Avedisian School of Medicine, Boston; Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston (Greenstein, Shih)
| | - Ryan Wang
- Departments of Psychiatry (Rivas-Grajales) and Neurology (Han, Wang), Boston University Chobanian & Avedisian School of Medicine, Boston; Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston (Greenstein, Shih)
| | - Patricia Greenstein
- Departments of Psychiatry (Rivas-Grajales) and Neurology (Han, Wang), Boston University Chobanian & Avedisian School of Medicine, Boston; Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston (Greenstein, Shih)
| | - Ludy C Shih
- Departments of Psychiatry (Rivas-Grajales) and Neurology (Han, Wang), Boston University Chobanian & Avedisian School of Medicine, Boston; Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston (Greenstein, Shih)
| |
Collapse
|
28
|
Liu T, Ren C, Guo W, Zhang X, Li Y, Wang Y, Zhang Q, Chen B, Dai J, Yan XX, Zhang J, Huo L, Cui M. Synthesis and preclinical evaluation of diarylamine derivative as Tau-PET radiotracer for Alzheimer's Disease. Eur J Med Chem 2025; 281:117046. [PMID: 39536496 DOI: 10.1016/j.ejmech.2024.117046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The presence of aggregated Tau in the brain is a dominant pathological hallmark of Tauopathies, particularly in Alzheimer's disease (AD). Therefore, developing ligands that can specifically and sensitively bind to Tau aggregates is essential for diagnosing and monitoring therapeutic interventions. In this study, we further investigated the structural optimization of the diarylamine skeleton, which exhibited promising binding characteristics and biological properties. We supplementarily explored the effects of the number and position of nitrogen atoms, types of heteroatoms and aromatic moieties, and radioactive positions on affinity for Tau. Through a structure-activity relationship (SAR) analysis based on 125I-labeled diarylamine derivatives, [125I]A6 was identified as a lead compound due to its desirable binding properties and ability to penetrate the brain, making it suitable for conversion into a18F-labeled PET tracer. Satisfactorily, [18F]FA1 fulfilled critical requirements as a Tau radiotracer, demonstrating high specificity and selectivity for Tau, a clean off-target profile against Aβ plaques and monoamine oxidase B (MAO-B), and favorable in vivo brain kinetics, as confirmed by dynamic PET studies in rodents and non-human primates.
Collapse
Affiliation(s)
- Tianqing Liu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Chao Ren
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Wantong Guo
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yuying Li
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China.
| | - Yan Wang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Hunan, 410013, China
| | - Qilei Zhang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Hunan, 410013, China
| | - Baian Chen
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China; Department of Laboratory Animal Sciences, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan, 430074, China.
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Hunan, 410013, China
| | - Jinming Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China.
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China; Center for Advanced Materials Research, Beijing Normal University at Zhuhai, Zhuhai, 519087, China.
| |
Collapse
|
29
|
Chunhui G, Yanqiu Y, Jibing C, Ning L, Fujun L. Exosomes and non-coding RNAs: bridging the gap in Alzheimer's pathogenesis and therapeutics. Metab Brain Dis 2025; 40:84. [PMID: 39754674 PMCID: PMC11700052 DOI: 10.1007/s11011-024-01520-7] [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] [Received: 08/30/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that primarily affects the elderly population and is the leading cause of dementia. Meanwhile, the vascular hypothesis suggests that vascular damage occurs in the early stages of the disease, leading to neurodegeneration and hindered waste clearance, which in turn triggers a series of events including the accumulation of amyloid plaques and Tau protein tangles. Non-coding RNAs (ncRNAs), including long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), have been found to be involved in the regulation of AD. Furthermore, lncRNAs and circRNAs can act as competitive endogenous RNAs to inhibit miRNAs, and their interactions can form a complex regulatory network. Exosomes, which are extracellular vesicles (EVs), are believed to be able to transfer ncRNAs between cells, thus playing a regulatory role in the brain by crossing the blood-brain barrier (BBB). Exosomes are part of the intercellular carrier system; therefore, utilizing exosomes to deliver drugs to recipient cells might not activate the immune system, making it a potential strategy to treat central nervous system diseases. In this review, we review that AD is a multifactorial neurological disease and that ncRNAs can regulate its multiple pathogenic mechanisms to improve our understanding of the etiology of AD and to simultaneously regulate multiple pathogenic mechanisms of AD through the binding of ncRNAs to exosomes to improve the treatment of AD.
Collapse
Affiliation(s)
- Guo Chunhui
- Graduate School, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - You Yanqiu
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Chen Jibing
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Luo Ning
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Li Fujun
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| |
Collapse
|
30
|
Șovrea AS, Boșca AB, Dronca E, Constantin AM, Crintea A, Suflețel R, Ștefan RA, Ștefan PA, Onofrei MM, Tschall C, Crivii CB. Non-Drug and Non-Invasive Therapeutic Options in Alzheimer's Disease. Biomedicines 2025; 13:84. [PMID: 39857667 PMCID: PMC11760896 DOI: 10.3390/biomedicines13010084] [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: 12/16/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Despite the massive efforts of modern medicine to stop the evolution of Alzheimer's disease (AD), it affects an increasing number of people, changing individual lives and imposing itself as a burden on families and the health systems. Considering that the vast majority of conventional drug therapies did not lead to the expected results, this review will discuss the newly developing therapies as an alternative in the effort to stop or slow AD. Focused Ultrasound (FUS) and its derived Transcranial Pulse Stimulation (TPS) are non-invasive therapeutic approaches. Singly or as an applied technique to change the permeability of the blood-brain-barrier (BBB), FUS and TPS have demonstrated the benefits of use in treating AD in animal and human studies. Adipose-derived stem Cells (ADSCs), gene therapy, and many other alternative methods (diet, sleep pattern, physical exercise, nanoparticle delivery) are also new potential treatments since multimodal approaches represent the modern trend in this disorder research therapies.
Collapse
Affiliation(s)
- Alina Simona Șovrea
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Adina Bianca Boșca
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Eleonora Dronca
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Anne-Marie Constantin
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Andreea Crintea
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Rada Suflețel
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Roxana Adelina Ștefan
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Paul Andrei Ștefan
- Radiology and Imaging Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania;
| | - Mădălin Mihai Onofrei
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Christoph Tschall
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Carmen-Bianca Crivii
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| |
Collapse
|
31
|
Rodríguez LC, Foressi NN, Celej MS. Liquid-liquid phase separation of tau and α-synuclein: A new pathway of overlapping neuropathologies. Biochem Biophys Res Commun 2024; 741:151053. [PMID: 39612640 DOI: 10.1016/j.bbrc.2024.151053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
Liquid-liquid phase separation (LLPS) is a critical phenomenon that leads to the formation of liquid-like membrane-less organelles within cells. Advances in our understanding of condensates reveal their significant roles in biology and highlight how their dysregulation may contribute to disease. Recent evidence indicates that the high protein concentration in coacervates may lead to abnormal protein aggregation associated with several neurodegenerative diseases. The presence of condensates containing multiple amyloidogenic proteins may play a role in the co-deposition and comorbidity seen in neurodegeneration. This review first provides a brief overview of the physicochemical bases and molecular determinants of LLPS. It then summarizes our understanding of Tau and α-synuclein (AS) phase separation, key proteins in Alzheimer's and Parkinson's diseases. By integrating recent findings on complex Tau and AS coacervation, this article offers a fresh perspective on how LLPS may contribute to the pathological overlap in neurodegenerative disorders and provide a novel therapeutic target to mitigate or prevent such conditions.
Collapse
Affiliation(s)
- Leandro Cruz Rodríguez
- Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Nahuel N Foressi
- Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - M Soledad Celej
- Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
32
|
Yoon S, Kim HY, Park S, Cha M, Kim K, Lee S, Kim J, Bhang S, Kim Y. Drug Discovery and Screening Tool Development for Tauopathies by Focusing on Pathogenic Tau Repeat 3 Oligomers. Angew Chem Int Ed Engl 2024; 63:e202411942. [PMID: 39314129 DOI: 10.1002/anie.202411942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
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.
Collapse
Affiliation(s)
- Soljee Yoon
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Hye Yun Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Sohui Park
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Minhae Cha
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Kyeonghwan Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Songmin Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - JiMin Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Saeyun Bhang
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
- Integrated Science and Engineering Division, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - YoungSoo Kim
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- POSTECH-Yonsei Campus, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- Amyloid Solution inc., 58 Pangyo-ro 255 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13486, Republic of Korea
| |
Collapse
|
33
|
Kumari S, Gupta S, Sukhija R, Gurjar S, Dubey SK, Taliyan R. Neuroprotective potential of Epigenetic modulators, its regulation and therapeutic approaches for the management of Parkinson's disease. Eur J Pharmacol 2024; 985:177123. [PMID: 39536854 DOI: 10.1016/j.ejphar.2024.177123] [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: 08/26/2024] [Revised: 10/19/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The progressive degeneration of dopaminergic neurons in the substantia nigra region of the brain leads to a deficiency of dopamine and, ultimately, the onset of Parkinson's disease (PD). Since there is currently no cure for PD, patients all around the world are dealing with symptomatic management. PD progression is influenced by multiple elements, such as environmental, biological, chemical, genetic, and epigenetic factors. Epigenetics is gaining increased attention due to its role in controlling the expression of genes that contribute to PD. Recent advancements in our understanding of the brain network and its related conditions have shown that alterations in gene expression may occur independently of genetic abnormalities. Therefore, a thorough investigation has been carried out to explore the significance of epigenetics in all degenerative disorders. Epigenetic modifications are essential for regulating cellular homeostasis. Therefore, a deeper understanding of these modifications might provide valuable insights into many diseases and potentially serve as targets for therapeutic interventions. This review article focuses on diverse epigenetic alterations linked to the progression of PD. These abnormalities are supported by numerous research on the control of gene expression and encompass all the epigenetic processes. The beginning of PD is intricately associated with aberrant DNA methylation mechanisms. DNA methyltransferases are the enzymes that create and preserve various DNA methylation patterns. Integrating epigenetic data with existing clinical methods for diagnosing PD may aid in discovering potential curative medicines and novel drug development approaches. This article solely addresses the importance of epigenetic modulators in PD, primarily the mechanisms of DNMTs, their roles in the development of PD, and their therapeutic approaches; it bypasses other PD therapies.
Collapse
Affiliation(s)
- Shobha Kumari
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India.
| | - Sakshi Gupta
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India.
| | - Rajesh Sukhija
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India.
| | - Shaifali Gurjar
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India.
| | | | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India.
| |
Collapse
|
34
|
Fare CM, Rothstein JD. Nuclear pore dysfunction and disease: a complex opportunity. Nucleus 2024; 15:2314297. [PMID: 38383349 PMCID: PMC10883112 DOI: 10.1080/19491034.2024.2314297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
The separation of genetic material from bulk cytoplasm has enabled the evolution of increasingly complex organisms, allowing for the development of sophisticated forms of life. However, this complexity has created new categories of dysfunction, including those related to the movement of material between cellular compartments. In eukaryotic cells, nucleocytoplasmic trafficking is a fundamental biological process, and cumulative disruptions to nuclear integrity and nucleocytoplasmic transport are detrimental to cell survival. This is particularly true in post-mitotic neurons, where nuclear pore injury and errors to nucleocytoplasmic trafficking are strongly associated with neurodegenerative disease. In this review, we summarize the current understanding of nuclear pore biology in physiological and pathological contexts and discuss potential therapeutic approaches for addressing nuclear pore injury and dysfunctional nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Charlotte M Fare
- Department of Neurology and Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey D Rothstein
- Department of Neurology and Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
35
|
McArthur N, Squire JD, Onyeachonam OJ, Bhatt NN, Jerez C, Holberton AL, Tessier PM, Wood LB, Kayed R, Kane RS. Generation of nanobodies with conformational specificity for tau oligomers that recognize tau aggregates from human Alzheimer's disease samples. Biomater Sci 2024; 12:6033-6046. [PMID: 39434503 PMCID: PMC11585960 DOI: 10.1039/d4bm00707g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024]
Abstract
Tauopathies are neurodegenerative diseases that involve tau misfolding and aggregation in the brain. These diseases, including Alzheimer's disease (AD), are some of the least understood and most difficult to treat neurodegenerative disorders. Antibodies and antibody fragments that target tau oligomers, which are especially toxic forms of tau, are promising options for immunotherapies and diagnostic tools for tauopathies. In this study, we have developed conformational, tau oligomer-specific nanobodies, or single-domain antibodies. We demonstrate that these nanobodies, OT2.4 and OT2.6, are highly specific for tau oligomers relative to tau monomers and fibrils. We used epitope mapping to verify that these nanobodies bind to discontinuous epitopes on tau and to support the idea that they interact with a conformation present in the oligomeric, and not monomeric or fibrillar, forms of tau. We show that these nanobodies interact with tau oligomers in brain samples from AD patients and from healthy older adults with primary age-related tauopathy. Our results demonstrate the potential of these nanobodies as tau oligomer-specific binding reagents and future tauopathy therapeutics and diagnostics.
Collapse
Affiliation(s)
- Nikki McArthur
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
| | - Jay D Squire
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
| | - Ogechukwu J Onyeachonam
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
| | - Nemil N Bhatt
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Abigail L Holberton
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
36
|
Tangavelou K, Bhaskar K. The Mechanistic Link Between Tau-Driven Proteotoxic Stress and Cellular Senescence in Alzheimer's Disease. Int J Mol Sci 2024; 25:12335. [PMID: 39596399 PMCID: PMC11595124 DOI: 10.3390/ijms252212335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
In Alzheimer's disease (AD), tau dissociates from microtubules (MTs) due to hyperphosphorylation and misfolding. It is degraded by various mechanisms, including the 20S proteasome, chaperone-mediated autophagy (CMA), 26S proteasome, macroautophagy, and aggrephagy. Neurofibrillary tangles (NFTs) form upon the impairment of aggrephagy, and eventually, the ubiquitin chaperone valosin-containing protein (VCP) and heat shock 70 kDa protein (HSP70) are recruited to the sites of NFTs for the extraction of tau for the ubiquitin-proteasome system (UPS)-mediated degradation. However, the impairment of tau degradation in neurons allows tau to be secreted into the extracellular space. Secreted tau can be monomers, oligomers, and paired helical filaments (PHFs), which are seeding competent pathological tau that can be endocytosed/phagocytosed by healthy neurons, microglia, astrocytes, oligodendrocyte progenitor cells (OPCs), and oligodendrocytes, often causing proteotoxic stress and eventually triggers senescence. Senescent cells secrete various senescence-associated secretory phenotype (SASP) factors, which trigger cellular atrophy, causing decreased brain volume in human AD. However, the molecular mechanisms of proteotoxic stress and cellular senescence are not entirely understood and are an emerging area of research. Therefore, this comprehensive review summarizes pertinent studies that provided evidence for the sequential tau degradation, failure, and the mechanistic link between tau-driven proteotoxic stress and cellular senescence in AD.
Collapse
Affiliation(s)
- Karthikeyan Tangavelou
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
37
|
Barker RM, Chambers A, Kehoe PG, Rowe E, Perks CM. Untangling the role of tau in sex hormone responsive cancers: lessons learnt from Alzheimer's disease. Clin Sci (Lond) 2024; 138:1357-1369. [PMID: 39469929 PMCID: PMC11522895 DOI: 10.1042/cs20230317] [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: 05/22/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Tubulin associated unit has been extensively studied in neurodegenerative diseases including Alzheimer's disease (AD), whereby its hyperphosphorylation and accumulation contributes to disease pathogenesis. Tau is abundantly expressed in the central nervous system but is also present in non-neuronal tissues and in tumours including sex hormone responsive cancers such as breast and prostate. Curiously, hormonal effects on tau also exist in an AD context from numerous studies on menopause, hormone replacement therapy, and androgen deprivation therapy. Despite sharing some risk factors, most importantly advancing age, there are numerous reports from population studies of, currently poorly explained inverse associations between cancer and Alzheimer's disease. We previously reviewed important components of the phosphoinositide-3-kinase/protein kinase B (PI3K/Akt) signalling pathway and their differential modulation in relation to the two diseases. Similarly, receptor tyrosine kinases, estrogen receptor and androgen receptor have all been implicated in the pathogenesis of both cancer and AD. In this review, we focus on tau and its effects in hormone responsive cancer in terms of development, progression, and treatment and in relation to sex hormones and PI3K/Akt signalling molecules including IRS-1, PTEN, Pin1, and p53.
Collapse
Affiliation(s)
- Rachel M. Barker
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Alfie Chambers
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Patrick G. Kehoe
- Department of Urology, Bristol Urological Institute, Southmead Hospital, Bristol BS10 5NB, UK
| | - Edward Rowe
- Dementia Research Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Claire M. Perks
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| |
Collapse
|
38
|
Su JF, Xiao Y, Wei LY, Lei HY, Sun F, Wang WX, Li SH, Wang XC, Zheng J, Wang JZ. A new tau dephosphorylation-targeting chimera for the treatment of tauopathies. Acta Pharmacol Sin 2024; 45:2267-2276. [PMID: 38956416 PMCID: PMC11489737 DOI: 10.1038/s41401-024-01326-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/26/2024] [Indexed: 07/04/2024]
Abstract
Abnormal accumulation of hyperphosphorylated tau protein plays a pivotal role in a collection of neurodegenerative diseases named tauopathies, including Alzheimer's disease (AD). We have recently conceptualized the design of hetero-bifunctional chimeras for selectively promoting the proximity between tau and phosphatase, thus specifically facilitating tau dephosphorylation and removal. Here, we sought to optimize the construction of tau dephosphorylating-targeting chimera (DEPTAC) and obtained a new chimera D14, which had high efficiency in reducing tau phosphorylation both in cell and tauopathy mouse models, while showing limited cytotoxicity. Moreover, D14 ameliorated neurodegeneration in primary cultured hippocampal neurons treated with toxic tau-K18 fragments, and improved cognitive functions of tauopathy mice. These results suggested D14 as a cost-effective drug candidate for the treatment of tauopathies.
Collapse
Affiliation(s)
- Jing-Fen Su
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Xiao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin-Yu Wei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hui-Yang Lei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fei Sun
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei-Xia Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shi-Hong Li
- Department of Anesthesiology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University; Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100083, China.
- Beijing Life Science Academy, Beijing, 102209, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
39
|
Kam MK, Park JY, Yun GH, Sohn HY, Park JH, Choi J, Koh YH, Jo C. Rottlerin Enhances the Autophagic Degradation of Phosphorylated Tau in Neuronal Cells. Mol Neurobiol 2024; 61:9633-9645. [PMID: 38671330 DOI: 10.1007/s12035-024-04182-9] [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: 11/29/2023] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
Intra-neuronal accumulation of hyper-phosphorylated tau as neurofibrillary tangles (NFT) is a hallmark of Alzheimer's disease (AD). To prevent the aggregation of phosphorylated tau in neurons, decreasing the phosphorylated tau protein levels is important. Here, we examined the biological effects of rottlerin, a phytochemical compound extracted from the Kamala tree, Mallotus philippinensis, on phosphorylated tau levels. Notably, rottlerin decreased the levels of intracellular phosphorylated and total tau. A marked increase in the LC3-II, a hallmark of autophagy, was observed in these cells, indicating that rottlerin strongly induced autophagy. Interestingly, rottlerin induced the phosphorylation of Raptor at S792 through the activation of adenosine-monophosphate activated-protein kinase (AMPK), which likely inhibits the mammalian target of rapamycin complex 1 (mTORC1), thus resulting in the activation of transcription factor EB (TFEB), a master regulator of autophagy. In addition, nuclear factor erythroid 2-related factor 2 (Nrf2) activity increased in the presence of rottlerin. The decrease of phosphorylated tau levels in the presence of rottlerin was ameliorated by the knockdown of TFEB and partially attenuated by the knockout of the Nrf2 gene. Taken together, rottlerin likely enhances the degradation of phosphorylated tau through autophagy activated by TFEB and Nrf2. Thus, our results suggest that a natural compound rottlerin could be used as a preventive and therapeutic drug for AD.
Collapse
Affiliation(s)
- Min Kyoung Kam
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Jee-Yun Park
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Gwang Ho Yun
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Hee-Young Sohn
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Jung Hyun Park
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Jiyoung Choi
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Young Ho Koh
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Chulman Jo
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea.
| |
Collapse
|
40
|
Gonzalez-Ortiz F, Karikari TK, Taylor-Te Vruchte D, Shepherd D, Kirsebom BE, Fladby T, Platt F, Blennow K. Plasma phosphorylated-tau217 is increased in Niemann-Pick disease type C. Brain Commun 2024; 6:fcae375. [PMID: 39502943 PMCID: PMC11535543 DOI: 10.1093/braincomms/fcae375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/15/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
Niemann-Pick disease type C and Alzheimer's disease are distinct neurodegenerative disorders that share the presence of neurofibrillary tangle pathology. In this multicentre study, we measured plasma phosphorylated-tau217 in controls (n = 60), Niemann-Pick disease type C (n = 71) and Alzheimer's disease (n = 30 positive for amyloid and negative for tau in CSF [A+T-] and n = 30 positive for both [A+T+]). Annual Severity Increment Score and Lysotracker measurements were evaluated in the Niemann-Pick disease type C group to estimate the rate of progression and lysosomal enlargement, respectively. In the cross-sectional analysis, plasma phosphorylated-tau217 was increased in Niemann-Pick disease type C compared with controls (2.52 ± 1.93 versus 1.02 ± 0.34 pg/mL, respectively, P < 0.001) and inversely correlated with age at disease onset (R = -0.54, P < 0.001). In the longitudinal analysis, plasma phosphorylated-tau217 was associated with disease progression determined by Annual Severity Increment Score (R = 0.48, P < 0.001) and lysosomal enlargement (R = 0.26, P = 0.004). We found no differences between A+T- Alzheimer's disease and Niemann-Pick disease type C (2.67 ± 1.18 versus 2.52 ± 1. 93 pg/mL, P = 0.31); however, A+T+ Alzheimer's disease had significantly higher levels than Niemann-Pick disease type C (3.26 ± 1.36 versus 2.52 ± 1.93 pg/mL, P = 0.001). Our findings suggest that plasma p-tau217 can increase in brain disorders with isolated tau pathology. Plasma p-tau217 associations with disease progression and severity make it a potential marker in Niemann-Pick disease type C.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 43180, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 43180, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 43180, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15203, USA
| | | | - Dawn Shepherd
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Bjørn-Eivind Kirsebom
- Department of Neurology, University Hospital of North Norway, Tromsø, 9019, Norway
- Department of Psychology, Faculty of Health Sciences, The Arctic University of Norway, Tromsø, 9031, Norway
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, 1478, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, 0316, Norway
| | - Frances Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 43180, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 43180, Sweden
- Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, 75013, France
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
41
|
Patel SP, Nikam T, Sreepathi B, Karankar VS, Jaiswal A, Vardhan SV, Rana A, Toga V, Srivastava N, Saraf SA, Awasthi S. Unraveling the Molecular Jam: How Crowding Shapes Protein Aggregation in Neurodegenerative Disorders. ACS Chem Biol 2024; 19:2118-2130. [PMID: 39373539 DOI: 10.1021/acschembio.4c00365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Protein misfolding and aggregation are the hallmarks of neurodegenerative diseases including Huntington's disease, Parkinson's disease, Alzheimer's disease, and prion diseases. A crowded cellular environment plays a crucial role in modulating protein aggregation processes in vivo and the pathological aggregation of proteins linked to different neurodegenerative disorders. Here, we review recent studies examining the effects of various crowding agents, such as polysaccharides, polyethylene glycol, and proteins like BSA and lysozyme on the behaviors of aggregation of several amyloidogenic peptides and proteins, including amylin, huntingtin, tau, α-synuclein, prion, and amyloid-β. We also summarize how the aggregation kinetics, thermodynamic stability, and morphology of amyloid fibrils are altered significantly in the presence of crowding agents. In addition, we also discuss the molecular basis underlying the modulation of amyloidogenic aggregation, focusing on changes in the protein conformation, and the nucleation mechanism. The molecular understanding of the effects of macromolecular crowding on amyloid aggregation is essential for revealing disease pathologies and identifying possible therapeutic targets. Thus, this review offers a perspective on the complex interplay between protein aggregation and the crowded cellular environment in vivo and explains the relevance of crowding in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Shashi Prakash Patel
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Tejas Nikam
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Bhargavi Sreepathi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Vijayshree S Karankar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Ankita Jaiswal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Salumuri Vamsi Vardhan
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Anika Rana
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Vanshu Toga
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Shubhini A Saraf
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Saurabh Awasthi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| |
Collapse
|
42
|
Li W, Dasgupta A, Yang K, Wang S, Hemandhar-Kumar N, Yarbro JM, Hu Z, Salovska B, Fornasiero EF, Peng J, Liu Y. An Extensive Atlas of Proteome and Phosphoproteome Turnover Across Mouse Tissues and Brain Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618303. [PMID: 39464138 PMCID: PMC11507808 DOI: 10.1101/2024.10.15.618303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Understanding how proteins in different mammalian tissues are regulated is central to biology. Protein abundance, turnover, and post-translational modifications like phosphorylation, are key factors that determine tissue-specific proteome properties. However, these properties are challenging to study across tissues and remain poorly understood. Here, we present Turnover-PPT, a comprehensive resource mapping the abundance and lifetime of 11,000 proteins and 40,000 phosphosites across eight mouse tissues and various brain regions, using advanced proteomics and stable isotope labeling. We revealed tissue-specific short- and long-lived proteins, strong correlations between interacting protein lifetimes, and distinct impacts of phosphorylation on protein turnover. Notably, we discovered that peroxisomes are regulated by protein turnover across tissues, and that phosphorylation regulates the stability of neurodegeneration-related proteins, such as Tau and α-synuclein. Thus, Turnover-PPT provides new fundamental insights into protein stability, tissue dynamic proteotypes, and the role of protein phosphorylation, and is accessible via an interactive web-based portal at https://yslproteomics.shinyapps.io/tissuePPT.
Collapse
Affiliation(s)
- Wenxue Li
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Abhijit Dasgupta
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Current address: Department of Computer Science and Engineering, SRM University AP, Neerukonda, Guntur, Andhra Pradesh 522240, India
| | - Ka Yang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Current address: Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Shisheng Wang
- Department of Pulmonary and Critical Care Medicine, and Proteomics-Metabolomics Analysis Platform, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Nisha Hemandhar-Kumar
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Jay M. Yarbro
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhenyi Hu
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
- Current address: Interdisciplinary Research center on Biology and chemistry, Shanghai institute of Organic chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Barbora Salovska
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Eugenio F. Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
- Department of Biomedical Informatics & Data Science, Yale University School of Medicine, New Haven, CT 06510, USA
- Lead Contact
| |
Collapse
|
43
|
Taniguchi D, Shimonaka S, Imtiaz A, Elahi M, Hatano T, Imai Y, Hattori N. Legumain/asparaginyl endopeptidase-resistant tau fibril fold produces corticobasal degeneration-specific C-terminal tau fragment. Neurobiol Dis 2024; 201:106686. [PMID: 39353514 DOI: 10.1016/j.nbd.2024.106686] [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: 06/06/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
Corticobasal degeneration (CBD) is a major four-repeat tauopathy along with progressive supranuclear palsy (PSP). Although detergent-insoluble 37-40-kDa carboxyl-terminal tau fragments (CTFs) are hallmarks of CBD pathology, the process of their formation is unknown. This study monitored the formation of CBD-type fibrils that exhibit astrocytic plaques, a characteristic CBD pathology, using its biochemical properties different from those of Alzheimer's disease/PSP-type fibrils. Tau fibrils from patients with CBD were amplified in non-astrocytic cultured cells, which maintained CBD-specific biochemical properties. We found that the lysosomal protease Legumain (LGMN) was involved in the generation of CBD-specific 37-40-kDa CTFs. While LGMN cleaved tau fibrils at Asn167 and Asn368 in the brain tissues of patients with Alzheimer's disease and PSP, tau fibrils from patients with CBD were predominantly resistant to cleavage at Asn368 by LGMN, resulting in the generation of CBD-specific CTFs. LGMN preference in tau fibrils was lost upon unraveling the tau fibril fold, suggesting that the CBD-specific tau fibril fold contributes to CBD-specific CTF production. From these findings, we found a way to differentiate astrocytic plaque from tufted astrocyte using the anti-Asn368 LGMN cleavage site-specific antibody. Inoculation of tau fibrils amplified in non-astrocytic cells into the mouse brain reproduced LGMN-resistant tau fibrils and recapitulated anti-Asn368-negative astrocytic plaques, which are characteristic of CBD pathology. This study supports the existence of disease-specific tau fibrils and contribute to further understanding of the tauopathy diagnosis. Our tau propagation mouse model using cellular tau seeds may contribute to uncovering disease mechanisms and screening for potential therapeutic compounds.
Collapse
Affiliation(s)
- Daisuke Taniguchi
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shotaro Shimonaka
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Ahmed Imtiaz
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Montasir Elahi
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yuzuru Imai
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan.
| |
Collapse
|
44
|
Doke R, Lamkhade GJ, Vinchurkar K, Singh S. Demystifying the Role of Neuroinflammatory Mediators as Biomarkers for Diagnosis, Prognosis, and Treatment of Alzheimer's Disease: A Review. ACS Pharmacol Transl Sci 2024; 7:2987-3003. [PMID: 39416969 PMCID: PMC11475310 DOI: 10.1021/acsptsci.4c00457] [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: 07/28/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Neuroinflammatory mediators play a pivotal role in the pathogenesis of Alzheimer's Disease (AD), influencing its onset, progression, and severity. The precise mechanisms behind AD are still not fully understood, leading current treatments to focus mainly on managing symptoms rather than preventing or curing the condition. The amyloid and tau hypotheses are the most widely accepted explanations for AD pathology; however, they do not completely account for the neuronal degeneration observed in AD. Growing evidence underscores the crucial role of neuroinflammation in the pathology of AD. The neuroinflammatory hypothesis presents a promising new approach to understanding the mechanisms driving AD. This review examines the importance of neuroinflammatory biomarkers in the diagnosis, prognosis, and treatment of AD. It delves into the mechanisms underlying neuroinflammation in AD, highlighting the involvement of various mediators such as cytokines, chemokines, and ROS. Additionally, this review discusses the potential of neuroinflammatory biomarkers as diagnostic tools, prognostic indicators, and therapeutic targets for AD management. By understanding the intricate interplay between neuroinflammation and AD pathology, this review aims to help in the development of efficient diagnostic and treatment plans to fight this debilitating neurological condition. Furthermore, it elaborates recent advancements in neuroimaging techniques and biofluid analysis for the identification and monitoring of neuroinflammatory biomarkers in AD patients.
Collapse
Affiliation(s)
- Rohit
R. Doke
- Jaihind
College of Pharmacy, Vadgaon Sahani, Pune, Maharashtra 412401, India
| | | | - Kuldeep Vinchurkar
- Krishna
School of Pharmacy, Kiran and Pallavi Patel
Global University, Vadodara, Gujarat 391243, India
| | - Sudarshan Singh
- Office
of Research Administration, Chiang Mai University, Chaing Mai 50200, Thailand
- Faculty
of Pharmacy, Chiang Mai University, Chaing Mai 50200, Thailand
| |
Collapse
|
45
|
Tripathi A, Pandey VK, Sharma G, Sharma AR, Taufeeq A, Jha AK, Kim JC. Genomic Insights into Dementia: Precision Medicine and the Impact of Gene-Environment Interaction. Aging Dis 2024; 15:2113-2135. [PMID: 38607741 PMCID: PMC11346410 DOI: 10.14336/ad.2024.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
The diagnosis, treatment, and management of dementia provide significant challenges due to its chronic cognitive impairment. The complexity of this condition is further highlighted by the impact of gene-environment interactions. A recent strategy combines advanced genomics and precision medicine methods to explore the complex genetic foundations of dementia. Utilizing the most recent research in the field of neurogenetics, the importance of precise genetic data in explaining the variation seen in dementia patients can be investigated. Gene-environment interactions are important because they influence genetic susceptibilities and aid in the development and progression of dementia. Modified to each patient's genetic profile, precision medicine has the potential to detect groups at risk and make previously unheard-of predictions about the course of diseases. Precision medicine techniques have the potential to completely transform treatment and diagnosis methods. Targeted medications that target genetic abnormalities will probably appear, providing the possibility for more efficient and customized medical interventions. Investigating the relationship between genes and the environment may lead to preventive measures that would enable people to change their surroundings and minimize the risk of dementia, leading to the improved lifestyle of affected people. This paper provides a comprehensive overview of the genomic insights into dementia, emphasizing the pivotal role of precision medicine, and gene-environment interactions.
Collapse
Affiliation(s)
- Anjali Tripathi
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Vinay Kumar Pandey
- Division of Research & Innovation (DRI), School of Applied & Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Garima Sharma
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Anam Taufeeq
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Kanpur, Uttar Pradesh, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Jin-Chul Kim
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
46
|
Goodman LD, Ralhan I, Li X, Lu S, Moulton MJ, Park YJ, Zhao P, Kanca O, Ghaderpour Taleghani ZS, Jacquemyn J, Shulman JM, Ando K, Sun K, Ioannou MS, Bellen HJ. Tau is required for glial lipid droplet formation and resistance to neuronal oxidative stress. Nat Neurosci 2024; 27:1918-1933. [PMID: 39187706 PMCID: PMC11809452 DOI: 10.1038/s41593-024-01740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 07/29/2024] [Indexed: 08/28/2024]
Abstract
The accumulation of reactive oxygen species (ROS) is a common feature of tauopathies, defined by Tau accumulations in neurons and glia. High ROS in neurons causes lipid production and the export of toxic peroxidated lipids (LPOs). Glia uptake these LPOs and incorporate them into lipid droplets (LDs) for storage and catabolism. We found that overexpressing Tau in glia disrupts LDs in flies and rat neuron-astrocyte co-cultures, sensitizing the glia to toxic, neuronal LPOs. Using a new fly tau loss-of-function allele and RNA-mediated interference, we found that endogenous Tau is required for glial LD formation and protection against neuronal LPOs. Similarly, endogenous Tau is required in rat astrocytes and human oligodendrocyte-like cells for LD formation and the breakdown of LPOs. Behaviorally, flies lacking glial Tau have decreased lifespans and motor defects that are rescuable by administering the antioxidant N-acetylcysteine amide. Overall, this work provides insights into the important role that Tau has in glia to mitigate ROS in the brain.
Collapse
Affiliation(s)
- Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Ye-Jin Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Program in Development, Disease Models and Therapeutics, Baylor College of Medicine, Houston, TX, USA
| | - Pinghan Zhao
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Ziyaneh S Ghaderpour Taleghani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Julie Jacquemyn
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Joshua M Shulman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Kanae Ando
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Program in Development, Disease Models and Therapeutics, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
47
|
Yang JY, Baek SE, Yoon JW, Kim HS, Kwon Y, Yeom E. Nesfatin-1 ameliorates pathological abnormalities in Drosophila hTau model of Alzheimer's disease. Biochem Biophys Res Commun 2024; 727:150311. [PMID: 38950494 DOI: 10.1016/j.bbrc.2024.150311] [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: 06/03/2024] [Revised: 06/07/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024]
Abstract
In human Alzheimer's disease (AD), the aggregation of tau protein is considered a significant hallmark, along with amyloid-beta. The formation of neurofibrillary tangles due to aberrant phosphorylation of tau disrupts microtubule stability, leading to neuronal toxicity, dysfunction, and subsequent cell death. Nesfatin-1 is a neuropeptide primarily known for regulating appetite and energy homeostasis. However, the function of Nesfatin-1 in a neuroprotective role has not been investigated. In this study, we aimed to elucidate the effect of Nesfatin-1 on tau pathology using the Drosophila model system. Our findings demonstrate that Nesfatin-1 effectively mitigates the pathological phenotypes observed in Drosophila human Tau overexpression models. Nesfatin-1 overexpression rescued the neurodegenerative phenotypes in the adult fly's eye and bristle. Additionally, Nesfatin-1 improved locomotive behavior, neuromuscular junction formation, and lifespan in the hTau AD model. Moreover, Nesfatin-1 controls tauopathy by reducing the protein level of hTau. Overall, this research highlights the potential therapeutic applications of Nesfatin-1 in ameliorating the pathological features associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Jae-Yoon Yang
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Si-Eun Baek
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Jong-Won Yoon
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Hyo-Sung Kim
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Younghwi Kwon
- KNU-G LAMP Project Group, KNU-Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Eunbyul Yeom
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; KNU-G LAMP Project Group, KNU-Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
48
|
Freitas DP, Saavedra J, Cardoso I, Gomes CM. Biophysical Studies of Amyloid-Binding Fluorophores to Tau AD Core Fibrils Formed without Cofactors. Int J Mol Sci 2024; 25:9946. [PMID: 39337433 PMCID: PMC11432123 DOI: 10.3390/ijms25189946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Tau is an intrinsically disordered protein involved in several neurodegenerative diseases where a common hallmark is the appearance of tau aggregates in the brain. One common approach to elucidate the mechanisms behind the aggregation of tau has been to recapitulate in vitro the self-assembly process in a fast and reproducible manner. While the seeding of tau aggregation is prompted by negatively charged cofactors, the obtained fibrils are morphologically distinct from those found in vivo. The Tau AD core fragment (TADC, tau 306-378) has emerged as a new model and potential solution for the cofactor-free in vitro aggregation of tau. Here, we use TADC to further study this process combining multiple amyloid-detecting fluorophores and fibril bioimaging. We confirmed by transmission electron microscopy that this fragment forms fibrils after quiescent incubation at 37 °C. We then employed a panel of eight amyloid-binding fluorophores to query the formed species by acquiring their emission spectra. The results obtained showed that nearly all dyes detect TADC self-assembled species. However, the successful monitoring of TADC aggregation kinetics was limited to three fluorophores (X-34, Bis-ANS, and pFTAA) which yielded sigmoidal curves but different aggregation half-times, hinting to different species being detected. Altogether, this study highlights the potential of using multiple extrinsic fluorescent probes, alone or in combination, as tools to further clarify mechanisms behind the aggregation of amyloidogenic proteins.
Collapse
Affiliation(s)
- Daniela P. Freitas
- BioISI—Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal;
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Joana Saavedra
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (J.S.); (I.C.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Isabel Cardoso
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (J.S.); (I.C.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Cláudio M. Gomes
- BioISI—Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal;
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| |
Collapse
|
49
|
Bhopatkar AA, Bhatt N, Haque MA, Xavier R, Fung L, Jerez C, Kayed R. MAPT mutations associated with familial tauopathies lead to formation of conformationally distinct oligomers that have cross-seeding ability. Protein Sci 2024; 33:e5099. [PMID: 39145409 PMCID: PMC11325167 DOI: 10.1002/pro.5099] [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: 04/21/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 08/16/2024]
Abstract
The microtubule associated protein, tau, is implicated in a multitude of neurodegenerative disorders that are collectively termed as tauopathies. These disorders are characterized by the presence of tau aggregates within the brain of afflicted individuals. Mutations within the MAPT gene that encodes the tau protein form the genetic backdrop for familial forms of tauopathies, such as frontotemporal dementia (FTD), but the molecular consequences of such alterations and their pathological effects are unclear. We sought to investigate the conformational properties of the aggregates of three tau mutants: A152T, P301L, and R406W, all implicated within FTD, and compare them to those of the native form (WT-Tau 2N4R). Our immunochemical analysis reveals that mutants and WT tau oligomers exhibit similar affinity for conformation-specific antibodies but have distinct morphology and secondary structure. Additionally, these oligomers possess different dye-binding properties and varying sensitivity to proteolytic processing. These results point to conformational variety among them. We then tested the ability of the mutant oligomers to cross-seed the aggregation of WT tau monomer. Using similar array of experiments, we found that cross-seeding with mutant aggregates leads to the formation of conformationally unique WT oligomers. The results discussed in this paper provide a novel perspective on the structural properties of oligomeric forms of WT tau 2N4R and its mutant, along with shedding some light on their cross-seeding behavior.
Collapse
Affiliation(s)
- Anukool A. Bhopatkar
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
- Present address:
Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Nemil Bhatt
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Md Anzarul Haque
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rhea Xavier
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Leiana Fung
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
- Present address:
Neuroscience Graduate Program, UT Southwestern Medical CenterDallasTexasUSA
| | - Cynthia Jerez
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
50
|
Wei YC, Kung YC, Lin C, Yeh CH, Chen PY, Huang WY, Shyu YC, Lin CP, Chen CK. Differential neuropsychiatric associations of plasma biomarkers in older adults with major depression and subjective cognitive decline. Transl Psychiatry 2024; 14:333. [PMID: 39152102 PMCID: PMC11329686 DOI: 10.1038/s41398-024-03049-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 06/23/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024] Open
Abstract
Older adults with major depressive disorder (MDD) or early cognitive decline during the subjective cognitive decline (SCD) stage may exhibit neuropsychiatric symptoms such as anxiety, depression, and subtle cognitive impairment. The clinicopathological features and biological mechanisms of MDD differ from those of SCD among older adults; these conditions thus require different treatment strategies. This study enrolled 82 participants above 50 years old with normal cognitive levels from the communities to examine biomarker-behavior correlations between MDD (n = 23) and SCD (n = 23) relative to a normal control (NC) group (n = 36). Multidomain assessments were performed for all participants, including immunomagnetic reduction tests to detect plasma beta-amyloid (Aβ), total tau (Tau), phosphorylated tau-181 (p-Tau181), neurofilament light chain, and glial fibrillary acidic protein (GFAP). This study observed that depressive symptoms in MDD were associated with amyloid pathology (plasma Aβ40 vs. HADS-D: R = 0.45, p = 0.031; Aβ42/Aβ40 vs. HADS-D: R = -0.47, p = 0.024), which was not observed in the NC (group difference p < 0.05). Moreover, cognitive decline in MDD was distinguished by a mixed neurodegenerative process involving amyloid (plasma Aβ42 vs. facial memory test: R = 0.48, p = 0.025), tau (Tau/Aβ42 vs. digit symbol substitution test (DSST): R = -0.53, p = 0.01), and astrocytic injury (plasma GFAP vs. Montreal cognitive assessment score: R = -0.44, p = 0.038; plasma GFAP vs. DSST: R = -0.52, p = 0.014), findings that did not apply to the NC (group difference p < 0.05). Moreover, this study revealed different biomarker-behavior correlations between individuals with SCD and the NC. Compared with the NC, cognitive decline in the SCD group might be unrelated to amyloid pathology and instead might be early manifestations of tau pathology. This study underscores the difference in clinicopathological features between MDD and SCD among older adults, which differ from those of the NC. These findings enhance our understanding of the mechanisms underlying MDD and SCD in older individuals.
Collapse
Affiliation(s)
- Yi-Chia Wei
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yi-Chia Kung
- Department of Radiology, Tri-Service General Hospital, Taipei, 114, Taiwan
| | - Chemin Lin
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Chun-Hung Yeh
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Pin-Yuan Chen
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Wen-Yi Huang
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yu-Chiau Shyu
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, 333, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Department of Education and Research, Taipei City Hospital, Taipei, 103, Taiwan.
| | - Chih-Ken Chen
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Psychiatry, Chang Gung Memorial Hospital, Keelung, 204, Taiwan.
| |
Collapse
|