1
|
Allahyartorkaman M, Chan TH, Chen EHL, Ng ST, Chen YA, Wen JK, Ho MR, Yen HY, Kuan YS, Kuo MH, Chen RPY. Phosphorylation-Induced Self-Coacervation versus RNA-Assisted Complex Coacervation of Tau Proteins. J Am Chem Soc 2025; 147:10172-10187. [PMID: 40074668 PMCID: PMC11951079 DOI: 10.1021/jacs.4c14728] [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: 10/21/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
In this study, the role of phosphorylation in the liquid-liquid phase separation (LLPS) of tau, the underlying driving forces, and the potential implications of this separation on protein conformation and subsequent protein aggregation were investigated. We compared in vivo-produced phosphorylated tau (p-tau) and nonphosphorylated tau under different coacervation conditions without adding crowding agents. Our findings revealed that spontaneous phase separation occurs exclusively in p-tau, triggered by a temperature shift from 4 °C to room temperature, and is driven by electrostatic and hydrophobic interactions. The p-tau self-acervation is reversible with temperature changes. Native mass spectrometry detects only two to nine phosphate groups per p-tau molecule, highlighting the impact of phosphorylation on tau's structural flexibility. Cross-linking mass spectrometry showed fewer long-range contacts in p-tau, suggesting a looser conformation induced by phosphorylation. Phosphorylation-induced LLPS and RNA-induced LLPS occurred at different timeframes. However, neither tau nor p-tau formed fibrils without the addition of dextran sulfate or RNA as inducers. Using human kidney epithelial cells expressing the tau R domain fused with fluorescent proteins as reporter cells, we observed aggregates in the nuclear envelope (NE) only in the cells treated with LLPS-state p-tau, which correlates with NE occurrences reported in Alzheimer's disease brain sections. These findings provide deeper insights into the impact of phosphorylation on tau aggregation through an intermediate condensation phase, offering novel perspectives on neurodegenerative disease mechanisms.
Collapse
Affiliation(s)
- Mohammadreza Allahyartorkaman
- Taiwan
International Graduate Program in Interdisciplinary Neuroscience, National Taiwan University and Academia Sinica, Taipei 115, Taiwan
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| | - Ting-Hsuan Chan
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Eric H.-L. Chen
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| | - See-Ting Ng
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| | - Yi-An Chen
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| | - Jung-Kun Wen
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| | - Meng-Ru Ho
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| | - Hsin-Yung Yen
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| | - Yung-Shu Kuan
- Institute
of Biochemical Sciences, National Taiwan
University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Min-Hao Kuo
- Department
of Biochemistry and Molecular Biology, Michigan
State University, 603 Wilson Road, Room 401, East Lansing, Michigan 48824, United States
| | - Rita P.-Y. Chen
- Institute
of Biological Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
- Neuroscience
Program of Academia Sinica, Academia Sinica, No. 128, Sec. 2, Academia Road,
Nankang, Taipei 115, Taiwan
| |
Collapse
|
2
|
Baldensperger T, Preissler M, Becker CFW. Non-enzymatic posttranslational protein modifications in protein aggregation and neurodegenerative diseases. RSC Chem Biol 2025; 6:129-149. [PMID: 39722676 PMCID: PMC11667106 DOI: 10.1039/d4cb00221k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Highly reactive metabolic intermediates and other small molecules frequently react with amino acid side chains, leading to non-enzymatic posttranslational modifications (nPTMs) of proteins. The abundance of these modifications increases under high metabolic activity or stress conditions and can dramatically impact protein structure and function. Although protein quality control mechanisms typically mitigate the effects of these impaired proteins, in long-lived and degradation-resistant proteins, nPTMs accumulate. In some cases, such as cataract development and diabetes, clear links between nPTMs, aging, and disease progression have been established. In neurodegenerative diseases such as Alzheimer's and Parkinson's disease, a key question is whether accumulation of nPTMs is a cause or consequence of protein aggregation. This review focuses on major nPTMs found on proteins with central roles in neurodegenerative diseases such as α-synuclein, β-amyloid, and tau. We summarize current knowledge on the formation of these modifications and discuss their potential impact on disease onset and progression. Additionally, we examine what is known to date about how nPTMs impair cellular detoxification, repair, and degradation systems. Finally, we critically discuss the available methodologies to systematically investigate nPTMs at the molecular level and outline suitable approaches to study their effects on protein aggregation. We aim to foster more research into the role of nPTMs in neurodegeneration by adapting methodologies that have proven successful in studying enzymatic posttranslational modifications. Specifically, we advocate for site-specific incorporation of these modifications into target proteins using advanced chemical and molecular biology techniques.
Collapse
Affiliation(s)
- Tim Baldensperger
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
| | - Miriam Preissler
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
- University of Vienna, Vienna Doctoral School in Chemistry (DoSChem) Währinger Str. 42 1090 Vienna Austria
| | - Christian F W Becker
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
| |
Collapse
|
3
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
4
|
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
|
5
|
Zhou F, Zhao Y, Sun Y, Chen W. Molecular Insights into Tau Pathology and its Therapeutic Strategies in Alzheimer's Disease. J Integr Neurosci 2024; 23:197. [PMID: 39613463 DOI: 10.31083/j.jin2311197] [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: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 12/01/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. The two major hallmarks of this disease are extracellular amyloid plaques and intracellular neurofibrillary tangles in the brain, accompanied by loss of neurons and synapses. The plaques and tangles mainly consist of amyloid-β (Aβ) and tau protein, respectively. Most of the therapeutic strategies for AD to date have focused on Aβ. However, there is still no effective therapy available. In recent years, the clinical therapeutic failure of targeting Aβ pathology has resulted in increased interest towards tau-based therapeutics. In the current review, we focus on the research progress regarding the pathological mechanisms of tau protein in this disease and discuss tau-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Futao Zhou
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yushi Zhao
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yangyan Sun
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Wanjiao Chen
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| |
Collapse
|
6
|
Chinnathambi S, Adithyan A, Suresh S, Velmurugan G, Chandrashekar M, Sahu S, Mishra M. Nuclear transport protein suppresses Tau neurodegeneration. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 143:363-385. [PMID: 39843141 DOI: 10.1016/bs.apcsb.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The nuclear pore complex, a large multimeric structure consists of numerous protein components, serves as a crucial gatekeeper for the transport of macromolecules across the nuclear envelope in eukaryotic cells. Dysfunction of the NPC has been implicated in various neurodegenerative diseases, including Alzheimer's disease. In AD, Tau aggregates interact with NPC proteins, known as nucleoporins, leading to disruptions in nuclear transport. Hyperphosphorylated Tau, a hallmark of AD pathology, interacts with central channel NUPs such as Nup62 and Nup98, causing cytoplasmic mis-localization of these proteins and impairing nuclear transport. Furthermore, Tau-NUP interactions promote Tau aggregation and the formation of neurofibrillary tangles, exacerbating neurodegeneration. Oligomeric Tau adheres to the lamin B receptor as well as nuclear lamin, preventing nucleocytoplasmic transport and resulting in heterochromatin unwinding, DNA damage, and neuronal death. The decrease in lamin B and increasing levels of lamin A along with C in AD-affected brain areas highlight the disease's intricacy. Furthermore, Tau internalization in the nucleus and interaction with nuclear pore complexes worsen NPC dysfunction, which contributes to neurotoxicity. Tau-DNA interactions suggest a chaperone-like role for Tau in DNA organization and repair, highlighting its involvement in maintaining genomic integrity. This review explores the intricate relationships between Tau, NPC components, and nuclear lamin in the context of AD, providing insights into the mechanisms underlying Tau-induced neurodegeneration and potential therapeutic targets.
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.
| | - Anusree Adithyan
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Swathi Suresh
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Gowshika Velmurugan
- 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
| | - Surajita Sahu
- Neural Development Biology Lab, Department of Life Science, NIT Rourkela, Rourkela, Odisha, India
| | - Monalisa Mishra
- Neural Development Biology Lab, Department of Life Science, NIT Rourkela, Rourkela, Odisha, India
| |
Collapse
|
7
|
Chinnathambi S, Velmurugan G, Suresh S, Adithyan A, Chandrashekar M. Nuclear Tau accumulation in Alzheimer's disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 143:323-337. [PMID: 39843139 DOI: 10.1016/bs.apcsb.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Tau is a well-known microtubule-associated protein and is located in the cytoplasm of neurons, which play a crucial role in Alzheimer's diseases. Due to its preferred binding to DNA sequences found in the nucleolus and pericentromeric heterochromatin, Tau has been found within the cell nucleus, where it may be a nucleic acid-associated protein. Tau has the ability to directly interact with nuclear pore complex nucleoporins, influencing both their structural and functional integrity. The interaction between Tau and NUPs highlights a potential mechanism underlying NPC dysfunction in AD pathogenesis. Pathological Tau hinders the import and export of nucleus through RAN mediated cascades. Nuclear Tau aggregates colocalize with membrane less organelles called nuclear speckles, which are involved in pre-mRNA splicing, and modify their dynamics, composition, and structure. Additionally, SRRM2 and other nuclear speckle proteins including MSUT2 and PABPN1 mislocalize to cytosolic Tau aggregates, and causes propagation of Tau aggregates. Research highlights, Extracellular Tau Oligomers induce significant nuclear invagination. They act as a key player in the transformation of healthy neurons into sick neurons in AD. The mechanism behind this phenomenon depends on intracellular Tau and is linked to changes in chromatin structure, nucleocytoplasmic transport, and gene transcription. This review highlights the vital roles of nuclear Tau protein in the context of nuclear pore complex functioning and, modulation of nuclear speckles in Alzheimer's diseases. Addressing these pathways is essential for formulating focused therapeutics intended to alleviate Tau-induced neurodegeneration.
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.
| | - Gowshika Velmurugan
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Swathi Suresh
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Anusree Adithyan
- 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
|
8
|
Rabanal-Ruiz Y, Pedrero-Prieto CM, Sanchez-Rodriguez L, Flores-Cuadrado A, Saiz-Sanchez D, Frontinan-Rubio J, Ubeda-Banon I, Duran Prado M, Martinez-Marcos A, Peinado JR. Differential accumulation of human β-amyloid and tau from enriched extracts in neuronal and endothelial cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167204. [PMID: 38679217 DOI: 10.1016/j.bbadis.2024.167204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
While Aβ and Tau cellular distribution has been largely studied, the comparative internalization and subcellular accumulation of Tau and Aβ isolated from human brain extracts in endothelial and neuronal cells has not yet been unveiled. We have previously demonstrated that controlled enrichment of Aβ from human brain extracts constitutes a valuable tool to monitor cellular internalization in vitro and in vivo. Herein, we establish an alternative method to strongly enrich Aβ and Tau aggregates from human AD brains, which has allowed us to study and compare the cellular internalization, distribution and toxicity of both proteins within brain barrier endothelial (bEnd.3) and neuronal (Neuro2A) cells. Our findings demonstrate the suitability of human enriched brain extracts to monitor the intracellular distribution of human Aβ and Tau, which, once internalized, show dissimilar sorting to different organelles within the cell and differential toxicity, exhibiting higher toxic effects on neuronal cells than on endothelial cells. While tau is strongly concentrated preferentially in mitochondria, Aβ is distributed predominantly within the endolysosomal system in endothelial cells, whereas the endoplasmic reticulum was its preferential location in neurons. Altogether, our findings display a picture of the interactions that human Aβ and Tau might establish in these cells.
Collapse
Affiliation(s)
- Y Rabanal-Ruiz
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - C M Pedrero-Prieto
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - L Sanchez-Rodriguez
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Flores-Cuadrado
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - D Saiz-Sanchez
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - J Frontinan-Rubio
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - I Ubeda-Banon
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - M Duran Prado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Martinez-Marcos
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Juan R Peinado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
9
|
Bukhari H, Nithianandam V, Battaglia RA, Cicalo A, Sarkar S, Comjean A, Hu Y, Leventhal MJ, Dong X, Feany MB. Transcriptional programs mediating neuronal toxicity and altered glial-neuronal signaling in a Drosophila knock-in tauopathy model. Genome Res 2024; 34:590-605. [PMID: 38599684 PMCID: PMC11146598 DOI: 10.1101/gr.278576.123] [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: 09/29/2023] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
Missense mutations in the gene encoding the microtubule-associated protein TAU (current and approved symbol is MAPT) cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human TAU in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas-mediated gene editing to model frontotemporal dementia caused by the TAU P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for Tau P251L display age-dependent neurodegeneration, display metabolic defects, and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies, we performed single-cell RNA sequencing on approximately 130,000 cells from brains of Tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified. Gene expression was also altered in glial cells, suggestive of non-cell-autonomous regulation. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell type-specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy that faithfully recapitulates the genetic context and phenotypic features of the human disease, and use the results of comprehensive single-cell sequencing analysis to outline pathways of neurotoxicity and highlight the potential role of non-cell-autonomous changes in glia.
Collapse
Affiliation(s)
- Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Vanitha Nithianandam
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Rachel A Battaglia
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Anthony Cicalo
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Matthew J Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, Massachusetts 02139, USA
| | - Xianjun Dong
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
10
|
Li S, Roy ER, Wang Y, Watkins T, Cao W. DLK-MAPK Signaling Coupled with DNA Damage Promotes Intrinsic Neurotoxicity Associated with Non-Mutated Tau. Mol Neurobiol 2024; 61:2978-2995. [PMID: 37955806 PMCID: PMC11043018 DOI: 10.1007/s12035-023-03720-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of neurodegeneration. Despite the well-established link between tau aggregation and clinical progression, the major pathways driven by this protein to intrinsically damage neurons are incompletely understood. To model AD-relevant neurodegeneration driven by tau, we overexpressed non-mutated human tau in primary mouse neurons and observed substantial axonal degeneration and cell death, a process accompanied by activated caspase 3. Mechanistically, we detected deformation of the nuclear envelope and increased DNA damage response in tau-expressing neurons. Gene profiling analysis further revealed significant alterations in the mitogen-activated protein kinase (MAPK) pathway; moreover, inhibitors of dual leucine zipper kinase (DLK) and c-Jun N-terminal kinase (JNK) were effective in alleviating wild-type human tau-induced neurodegeneration. In contrast, mutant P301L human tau was less toxic to neurons, despite causing comparable DNA damage. Axonal DLK activation induced by wild-type tau potentiated the impact of DNA damage response, resulting in overt neurotoxicity. In summary, we have established a cellular tauopathy model highly relevant to AD and identified a functional synergy between the DLK-MAPK axis and DNA damage response in the neuronal degenerative process.
Collapse
Affiliation(s)
- Sanming Li
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Ethan R Roy
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yanyu Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Trent Watkins
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Wei Cao
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
Datta D, Perone I, Wijegunawardana D, Liang F, Morozov YM, Arellano J, Duque A, Xie Z, van Dyck CH, Joyce MKP, Arnsten AFT. Nanoscale imaging of pT217-tau in aged rhesus macaque entorhinal and dorsolateral prefrontal cortex: Evidence of interneuronal trafficking and early-stage neurodegeneration. Alzheimers Dement 2024; 20:2843-2860. [PMID: 38445818 PMCID: PMC11032534 DOI: 10.1002/alz.13737] [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: 10/06/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Tau phosphorylated at threonine-217 (pT217-tau) is a novel fluid-based biomarker that predicts onset of Alzheimer's disease (AD) symptoms, but little is known about how pT217-tau arises in the brain, as soluble pT217-tau is dephosphorylated post mortem in humans. METHODS We used multilabel immunofluorescence and immunoelectron microscopy to examine the subcellular localization of early-stage pT217-tau in entorhinal and prefrontal cortices of aged macaques with naturally occurring tau pathology and assayed pT217-tau levels in plasma. RESULTS pT217-tau was aggregated on microtubules within dendrites exhibiting early signs of degeneration, including autophagic vacuoles. It was also seen trafficking between excitatory neurons within synapses on spines, where it was exposed to the extracellular space, and thus accessible to cerebrospinal fluid (CSF)/blood. Plasma pT217-tau levels increased across the age span and thus can serve as a biomarker in macaques. DISCUSSION These data help to explain why pT217-tau predicts degeneration in AD and how it gains access to CSF and plasma to serve as a fluid biomarker.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
- Department of PsychiatryYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Isabella Perone
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | | | - Feng Liang
- Department of AnesthesiaCritical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Yury M. Morozov
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Jon Arellano
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Alvaro Duque
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Zhongcong Xie
- Department of AnesthesiaCritical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Mary Kate P. Joyce
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| | - Amy F. T. Arnsten
- Department of NeuroscienceYale UniversitySchool of MedicineNew HavenConnecticutUSA
| |
Collapse
|
12
|
Sexton CE, Bitan G, Bowles KR, Brys M, Buée L, Maina MB, Clelland CD, Cohen AD, Crary JF, Dage JL, Diaz K, Frost B, Gan L, Goate AM, Golbe LI, Hansson O, Karch CM, Kolb HC, La Joie R, Lee SE, Matallana D, Miller BL, Onyike CU, Quiroz YT, Rexach JE, Rohrer JD, Rommel A, Sadri‐Vakili G, Schindler SE, Schneider JA, Sperling RA, Teunissen CE, Weninger SC, Worley SL, Zheng H, Carrillo MC. Novel avenues of tau research. Alzheimers Dement 2024; 20:2240-2261. [PMID: 38170841 PMCID: PMC10984447 DOI: 10.1002/alz.13533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 01/05/2024]
Abstract
INTRODUCTION The pace of innovation has accelerated in virtually every area of tau research in just the past few years. METHODS In February 2022, leading international tau experts convened to share selected highlights of this work during Tau 2022, the second international tau conference co-organized and co-sponsored by the Alzheimer's Association, CurePSP, and the Rainwater Charitable Foundation. RESULTS Representing academia, industry, and the philanthropic sector, presenters joined more than 1700 registered attendees from 59 countries, spanning six continents, to share recent advances and exciting new directions in tau research. DISCUSSION The virtual meeting provided an opportunity to foster cross-sector collaboration and partnerships as well as a forum for updating colleagues on research-advancing tools and programs that are steadily moving the field forward.
Collapse
Affiliation(s)
| | - Gal Bitan
- Department of NeurologyDavid Geffen School of MedicineBrain Research InstituteMolecular Biology InstituteUniversity of California Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Kathryn R. Bowles
- UK Dementia Research Institute at the University of EdinburghCentre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | | | - Luc Buée
- Univ LilleInsermCHU‐LilleLille Neuroscience and CognitionLabEx DISTALZPlace de VerdunLilleFrance
| | - Mahmoud Bukar Maina
- Sussex NeuroscienceSchool of Life SciencesUniversity of SussexFalmerUK
- Biomedical Science Research and Training CentreYobe State UniversityDamaturuNigeria
| | - Claire D. Clelland
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ann D. Cohen
- University of PittsburghSchool of MedicineDepartment of Psychiatry and Alzheimer's disease Research CenterPittsburghPennsylvaniaUSA
| | - John F. Crary
- Departments of PathologyNeuroscience, and Artificial Intelligence & Human HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jeffrey L. Dage
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Bess Frost
- Sam & Ann Barshop Institute for Longevity & Aging Studies Glenn Biggs Institute for Alzheimer's & Neurodegenerative Disorders Department of Cell Systems and Anatomy University of Texas Health San AntonioSan AntonioTexasUSA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research InstituteFeil Family Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Alison M Goate
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's diseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Lawrence I. Golbe
- CurePSPIncNew YorkNew YorkUSA
- Rutgers Robert Wood Johnson Medical SchoolNew BrunswickNew JerseyUSA
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöLund UniversityLundSweden
| | - Celeste M. Karch
- Department of PsychiatryWashington University in St. LouisSt. LouisMissouriUSA
| | | | - Renaud La Joie
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Suzee E. Lee
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Diana Matallana
- Aging InstituteNeuroscience ProgramPsychiatry DepartmentSchool of MedicinePontificia Universidad JaverianaBogotáColombia
- Mental Health DepartmentHospital Universitario Fundaciòn Santa FeBogotaColombia
| | - Bruce L. Miller
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Chiadi U. Onyike
- Division of Geriatric Psychiatry and NeuropsychiatryJohns Hopkins University School of MedicineBaltimoreBaltimoreMarylandUSA
| | - Yakeel T. Quiroz
- Departments of Psychiatry and NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Jessica E. Rexach
- Program in NeurogeneticsDepartment of NeurologyDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Jonathan D. Rohrer
- Department of Neurodegenerative DiseaseDementia Research CentreUniversity College London Institute of Neurology, Queen SquareLondonUK
| | - Amy Rommel
- Rainwater Charitable FoundationFort WorthTexasUSA
| | - Ghazaleh Sadri‐Vakili
- Sean M. Healey &AMG Center for ALS at Mass GeneralMassachusetts General HospitalBostonMassachusettsUSA
| | - Suzanne E. Schindler
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Reisa A. Sperling
- Center for Alzheimer Research and TreatmentBrigham and Women's HospitalMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryClinical Chemistry departmentAmsterdam NeuroscienceProgram NeurodegenerationAmsterdam University Medical CentersVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | | | | | - Hui Zheng
- Huffington Center on AgingBaylor College of MedicineHoustonTexasUSA
| | | |
Collapse
|
13
|
Bukhari H, Nithianandam V, Battaglia RA, Cicalo A, Sarkar S, Comjean A, Hu Y, Leventhal MJ, Dong X, Feany MB. Transcriptional programs mediating neuronal toxicity and altered glial-neuronal signaling in a Drosophila knock-in tauopathy model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578624. [PMID: 38352559 PMCID: PMC10862891 DOI: 10.1101/2024.02.02.578624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Missense mutations in the gene encoding the microtubule-associated protein tau cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human tau in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease, but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas mediated gene editing to model frontotemporal dementia caused by the tau P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for tau P251L display age-dependent neurodegeneration, metabolic defects and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies we performed single-cell RNA sequencing on approximately 130,000 cells from brains of tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified and non-cell autonomously in glial cells. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell-type specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy, which faithfully recapitulates the genetic context and phenotypic features of the human disease and use the results of comprehensive single cell sequencing analysis to outline pathways of neurotoxicity and highlight the role of non-cell autonomous changes in glia.
Collapse
Affiliation(s)
- Hassan Bukhari
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Vanitha Nithianandam
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Rachel A. Battaglia
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Anthony Cicalo
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Genomics and Bioinformatics Hub, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Matthew J. Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA 02139
| | - Xianjun Dong
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Genomics and Bioinformatics Hub, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Mel B. Feany
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| |
Collapse
|
14
|
Singh N, Sharma S, Ghosh KK, Gupta B, Kuca K. Prominent Perspective on Existing Biological Hallmarks of Alzheimer's Disease. Curr Top Med Chem 2024; 24:1120-1133. [PMID: 38591203 DOI: 10.2174/0115680266292514240404040341] [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: 11/12/2023] [Revised: 02/24/2024] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
Biomarkers are the most significant diagnosis tools tending towards unique approaches and solutions for the prevention and cure of Alzheimer's Disease (AD). The current report provides a clear perception of the concept of various biomarkers and their prominent features through analysis to provide a possible solution for the inhibition of events in AD. Scientists around the world truly believe that crucial hallmarks can serve as critical tools in the early diagnosis, cure, and prevention, as well as the future of medicine. The awareness and understanding of such biomarkers would provide solutions to the puzzled mechanism of this neuronal disorder. Some of the argued biomarkers in the present article are still in an experimental phase as they need to undergo specific clinical trials before they can be considered for treatment.
Collapse
Affiliation(s)
- Namrata Singh
- Department of Engineering Science, Ramrao Adik Institute of Technology, DY Patil University, Navi Mumbai, 400706, India
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Srishti Sharma
- School of Studies in Chemistry, Pt. Ravishankar Shukla University, Raipur, 492010 (C.G.), India
| | - Kallol K Ghosh
- School of Studies in Chemistry, Pt. Ravishankar Shukla University, Raipur, 492010 (C.G.), India
| | - Bhanushree Gupta
- Centre of Basic Sciences, Pt. Ravishankar Shukla University, Raipur, 492010 (C.G.), India
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
- Research Institute for Biomedical Science, University of Hradec Králové, Antonína Dvoraka 451/1, 500 02 Hradec Kralove, Czech Republic
| |
Collapse
|
15
|
Alonso ADC, El Idrissi A, Candia R, Morozova V, Kleiman FE. Tau: More than a microtubule-binding protein in neurons. Cytoskeleton (Hoboken) 2024; 81:71-77. [PMID: 37819542 DOI: 10.1002/cm.21795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Tau protein was discovered as a microtubule-associated protein nearly 50 years ago, and our understanding of tau has revolved around that role. Even with tau's rise to stardom as a central player in neurodegenerative disease, therapeutic efforts have largely been targeted toward cytoskeletal changes. While some studies hinted toward non-cytoskeletal roles for tau, it is only fairly recently that these ideas have begun to receive considerable attention. Many new binding partners for tau have been identified, including DNA, RNA, RNA-binding proteins, some receptors, and other tau molecules. The diversity of tau binding partners coupled with the discovery of tau other than axonal compartments such as nucleus, dendrites, and synapses have led to the proposal of novel functions for tau in roles such as nuclear stability, cell signaling, transcriptional processing, and protein synthesis. Tau self-assembly in particular has made an impact, leading to the hypothesis that a prion-like function of hyperphosphorylated tau is central to tauopathies. With tau emerging as a multifaceted protein that operates in many parts of the cell and with many molecular partners, the field of tau biology is primed for discoveries that can provide new perspectives on both the unique biochemistry of tau and the nature of devastating neurological diseases.
Collapse
Affiliation(s)
- Alejandra Del Carmen Alonso
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Abdeslem El Idrissi
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Robert Candia
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Viktoriya Morozova
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
- Helene Fuld College of Nursing, New York, New York, USA
| | - Frida Esther Kleiman
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
- Chemistry Department, Hunter College, The City University of New York, New York, New York, USA
| |
Collapse
|
16
|
Portillo M, Toiber D. Tau's function and dysfunction in the brain: when small changes have big consequences. Neural Regen Res 2024; 19:152-153. [PMID: 37488858 PMCID: PMC10479848 DOI: 10.4103/1673-5374.373682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/14/2023] [Accepted: 03/26/2023] [Indexed: 07/26/2023] Open
Affiliation(s)
- Miguel Portillo
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Debra Toiber
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
17
|
Younas N, Saleem T, Younas A, Zerr I. Nuclear face of Tau: an inside player in neurodegeneration. Acta Neuropathol Commun 2023; 11:196. [PMID: 38087392 PMCID: PMC10714511 DOI: 10.1186/s40478-023-01702-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Tau (Tubulin associated unit) protein is a major hallmark of Alzheimer's disease (AD) and tauopathies. Tau is predominantly an axonal protein with a crucial role in the stabilization and dynamics of the microtubules. Since the discovery of Tau protein in 1975, research efforts were concentrated on the pathophysiological role of Tau protein in the context of the microtubules. Although, for more than three decades, different localizations of Tau protein have been discovered e.g., in the nuclear compartments. Discovery of the role of Tau protein in various cellular compartments especially in the nucleus opens up a new fold of complexity in tauopathies. Data from cellular models, animal models, and the human brain indicate that nuclear Tau is crucial for genome stability and to cope with cellular distress. Moreover, it's nature of nuclear translocation, its interactions with the nuclear DNA/RNA and proteins suggest it could play multiple roles in the nucleus. To comprehend Tau pathophysiology and efficient Tau-based therapies, there is an urgent need to understand whole repertoire of Tau species (nuclear and cytoplasmic) and their functional relevance. To complete the map of Tau repertoire, understanding of various species of Tau in the nucleus and cytoplasm, identification if specific transcripts of Tau, isoforms and post-translational modifications could foretell Tau's localizations and functions, and how they are modified in neurodegenerative diseases like AD, is urgently required. In this review, we explore the nuclear face of Tau protein, its nuclear localizations and functions and its linkage with Alzheimer's disease.
Collapse
Affiliation(s)
- Neelam Younas
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany.
| | - Tayyaba Saleem
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany
| | - Abrar Younas
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany
| | - Inga Zerr
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany
| |
Collapse
|
18
|
Olopade JO, Mustapha OA, Fatola OI, Ighorodje E, Folarin OR, Olopade FE, Omile IC, Obasa AA, Oyagbemi AA, Olude MA, Thackray AM, Bujdoso R. Neuropathological profile of the African Giant Rat brain (Cricetomys gambianus) after natural exposure to heavy metal environmental pollution in the Nigerian Niger Delta. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:120496-120514. [PMID: 37945948 DOI: 10.1007/s11356-023-30619-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023]
Abstract
Pollution by heavy metals is a threat to public health because of the adverse effects on multiple organ systems including the brain. Here, we used the African giant rat (AGR) as a novel sentinel host to assess the effect of heavy metal accumulation and consequential neuropathology upon the brain. For this study, AGR were collected from distinct geographical regions of Nigeria: the rain forest region of south-west Nigeria (Ibadan), the central north of Nigeria (Abuja), and in oil-polluted areas of south Nigeria (Port-Harcourt). We found that zinc, copper, and iron were the major heavy metals that accumulated in the brain and serum of sentinel AGR, with the level of iron highest in animals from Port-Harcourt and least in animals from Abuja. Brain pathology, determined by immunohistochemistry markers of inflammation and oxidative stress, was most severe in animals from Port Harcourt followed by those from Abuja and those from Ibadan were the least affected. The brain pathologies were characterized by elevated brain advanced oxidation protein product (AOPP) levels, neuronal depletion in the prefrontal cortex, severe reactive astrogliosis in the hippocampus and cerebellar white matter, demyelination in the subcortical white matter and cerebellar white matter, and tauopathies. Selective vulnerabilities of different brain regions to heavy metal pollution in the AGR collected from the different regions of the country were evident. In conclusion, we propose that neuropathologies associated with redox dyshomeostasis because of environmental pollution may be localized and contextual, even in a heavily polluted environment. This novel study also highlights African giant rats as suitable epidemiological sentinels for use in ecotoxicological studies.
Collapse
Affiliation(s)
- James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria.
| | - Oluwaseun Ahmed Mustapha
- Neuroscience Unit, Department of Veterinary Anatomy, College of Veterinary Medicine, Federal University of Agriculture Abeokuta, Abeokuta, Ogun State, Nigeria
| | - Olanrewaju Ifeoluwa Fatola
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Ejiro Ighorodje
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Oluwabusayo Racheal Folarin
- Department of Biomedical Laboratory Science, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | | | - Irene Chizubelu Omile
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Adedunsola Ajike Obasa
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Matthew Ayokunle Olude
- Neuroscience Unit, Department of Veterinary Anatomy, College of Veterinary Medicine, Federal University of Agriculture Abeokuta, Abeokuta, Ogun State, Nigeria
| | - Alana Maureen Thackray
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Raymond Bujdoso
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| |
Collapse
|
19
|
Datta D, Perone I, Wijegunawardana D, Liang F, Morozov YM, Arellano J, Duque A, Xie Z, van Dyck CH, Arnsten AFT. Nanoscale imaging of pT217-tau in aged rhesus macaque entorhinal and dorsolateral prefrontal cortex: Evidence of interneuronal trafficking and early-stage neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566046. [PMID: 37986900 PMCID: PMC10659394 DOI: 10.1101/2023.11.07.566046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
INTRODUCTION pT217-tau is a novel fluid-based biomarker that predicts onset of Alzheimer's disease (AD) symptoms, but little is known about how pT217-tau arises in brain, as soluble pT217-tau is dephosphorylated postmortem in humans. METHODS We utilized multi-label immunofluorescence and immunoelectron-microscopy to examine the subcellular localization of early-stage pT217-tau in entorhinal and prefrontal cortices of aged macaques with naturally-occurring tau pathology and assayed pT217-tau levels in plasma. RESULTS pT217-tau was aggregated on microtubules within dendrites exhibiting early signs of degeneration, including autophagic vacuoles. It was also seen trafficking between excitatory neurons within synapses on spines, where it was exposed to the extracellular space, and thus accessible to CSF/blood. Plasma pT217-tau levels increased across the age-span and thus can serve as a biomarker in macaques. DISCUSSION These data help to explain why pT217-tau predicts degeneration in AD and how it gains access to CSF and plasma to serve as a fluid biomarker.
Collapse
|
20
|
Sturiale V, Bruno F, Brancato D, D’Amico AG, Maugeri G, D’Agata V, Saccone S, Federico C. Cell Cycle Reactivation, at the Start of Neurodegeneration, Induced by Forskolin and Aniline in Differentiated Neuroblastoma Cells. Int J Mol Sci 2023; 24:14373. [PMID: 37762676 PMCID: PMC10531780 DOI: 10.3390/ijms241814373] [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: 08/28/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
A characteristic hallmark of Alzheimer's disease (AD) is the intracellular accumulation of hyperphosphorylated tau protein, a phenomenon that appears to have associations with oxidative stress, double-stranded DNA breakage, and the de-condensation of heterochromatin. Re-entry into the cell division cycle appears to be involved in the onset of this neurodegenerative process. Indeed, the cell cycle cannot proceed regularly in the differentiated neurons leading to cell death. Here, we induced cell cycle reactivation in neuronal-like cells, obtained by neuroblastoma cells treated with retinoic acid, by exposure to forskolin or aniline. These compounds determine tau hyperphosphorylation or oxidative stress, respectively, resulting in the appearance of features resembling the start of neuronal degeneration typical of AD, such as tau hyperphosphorylation and re-entry into the cell cycle. Indeed, we detected an increased transcriptional level of cyclins and the appearance of a high number of mitotic cells. We also observed a delay in the initiation of the cell cycle when forskolin was co-administered with pituitary adenylate cyclase-activating polypeptide (PACAP). This delay was not observed when PACAP was co-administered with aniline. Our data demonstrate the relevance of tau hyperphosphorylation in initiating an ectopic cell cycle in differentiated neuronal cells, a condition that can lead to neurodegeneration. Moreover, we highlight the utility of neuroblastoma cell lines as an in vitro cellular model to test the possible neuroprotective effects of natural molecules.
Collapse
Affiliation(s)
- Valentina Sturiale
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Francesca Bruno
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Desiree Brancato
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Agata Grazia D’Amico
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Velia D’Agata
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Concetta Federico
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| |
Collapse
|
21
|
Morello G, Guarnaccia M, La Cognata V, Latina V, Calissano P, Amadoro G, Cavallaro S. Transcriptomic Analysis in the Hippocampus and Retina of Tg2576 AD Mice Reveals Defective Mitochondrial Oxidative Phosphorylation and Recovery by Tau 12A12mAb Treatment. Cells 2023; 12:2254. [PMID: 37759477 PMCID: PMC10527038 DOI: 10.3390/cells12182254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Increasing evidence implicates decreased energy metabolism and mitochondrial dysfunctions among the earliest pathogenic events of Alzheimer's disease (AD). However, the molecular mechanisms underlying bioenergetic dysfunctions in AD remain, to date, largely unknown. In this work, we analyzed transcriptomic changes occurring in the hippocampus and retina of a Tg2576 AD mouse model and wild-type controls, evaluating their functional implications by gene set enrichment analysis. The results revealed that oxidative phosphorylation and mitochondrial-related pathways are significantly down-regulated in both tissues of Tg2576 mice, supporting the role of these processes in the pathogenesis of AD. In addition, we also analyzed transcriptomic changes occurring in Tg2576 mice treated with the 12A12 monoclonal antibody that neutralizes an AD-relevant tau-derived neurotoxic peptide in vivo. Our analysis showed that the mitochondrial alterations observed in AD mice were significantly reverted by treatment with 12A12mAb, supporting bioenergetic pathways as key mediators of its in vivo neuroprotective and anti-amyloidogenic effects. This study provides, for the first time, a comprehensive characterization of molecular events underlying the disrupted mitochondrial bioenergetics in AD pathology, laying the foundation for the future development of diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Giovanna Morello
- Institute for Biomedical Research and Innovation, National Research Council (CNR-IRIB), Via Paolo Gaifami, 18, 95126 Catania, Italy; (G.M.); (M.G.); (V.L.C.)
| | - Maria Guarnaccia
- Institute for Biomedical Research and Innovation, National Research Council (CNR-IRIB), Via Paolo Gaifami, 18, 95126 Catania, Italy; (G.M.); (M.G.); (V.L.C.)
| | - Valentina La Cognata
- Institute for Biomedical Research and Innovation, National Research Council (CNR-IRIB), Via Paolo Gaifami, 18, 95126 Catania, Italy; (G.M.); (M.G.); (V.L.C.)
| | - Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy; (V.L.); (P.C.); (G.A.)
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy; (V.L.); (P.C.); (G.A.)
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy; (V.L.); (P.C.); (G.A.)
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council (CNR-IRIB), Via Paolo Gaifami, 18, 95126 Catania, Italy; (G.M.); (M.G.); (V.L.C.)
| |
Collapse
|
22
|
Daniilidou M, Eroli F, Alanko V, Goikolea J, Latorre-Leal M, Rodriguez-Rodriguez P, Griffiths WJ, Wang Y, Pacciarini M, Brinkmalm A, Zetterberg H, Blennow K, Rosenberg A, Bogdanovic N, Winblad B, Kivipelto M, Ibghi D, Cedazo-Minguez A, Maioli S, Matton A. Alzheimer's disease biomarker profiling in a memory clinic cohort without common comorbidities. Brain Commun 2023; 5:fcad228. [PMID: 37680670 PMCID: PMC10481253 DOI: 10.1093/braincomms/fcad228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/17/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Alzheimer's disease is a multifactorial disorder with large heterogeneity. Comorbidities such as hypertension, hypercholesterolaemia and diabetes are known contributors to disease progression. However, less is known about their mechanistic contribution to Alzheimer's pathology and neurodegeneration. The aim of this study was to investigate the relationship of several biomarkers related to risk mechanisms in Alzheimer's disease with the well-established Alzheimer's disease markers in a memory clinic population without common comorbidities. We investigated 13 molecular markers representing key mechanisms underlying Alzheimer's disease pathogenesis in CSF from memory clinic patients without diagnosed hypertension, hypercholesterolaemia or diabetes nor other neurodegenerative disorders. An analysis of covariance was used to compare biomarker levels between clinical groups. Associations were analysed by linear regression. Two-step cluster analysis was used to determine patient clusters. Two key markers were analysed by immunofluorescence staining in the hippocampus of non-demented control and Alzheimer's disease individuals. CSF samples from a total of 90 participants were included in this study: 30 from patients with subjective cognitive decline (age 62.4 ± 4.38, female 60%), 30 with mild cognitive impairment (age 65.6 ± 7.48, female 50%) and 30 with Alzheimer's disease (age 68.2 ± 7.86, female 50%). Angiotensinogen, thioredoxin-1 and interleukin-15 had the most prominent associations with Alzheimer's disease pathology, synaptic and axonal damage markers. Synaptosomal-associated protein 25 kDa and neurofilament light chain were increased in mild cognitive impairment and Alzheimer's disease patients. Grouping biomarkers by biological function showed that inflammatory and survival components were associated with Alzheimer's disease pathology, synaptic dysfunction and axonal damage. Moreover, a vascular/metabolic component was associated with synaptic dysfunction. In the data-driven analysis, two patient clusters were identified: Cluster 1 had increased CSF markers of oxidative stress, vascular pathology and neuroinflammation and was characterized by elevated synaptic and axonal damage, compared with Cluster 2. Clinical groups were evenly distributed between the clusters. An analysis of post-mortem hippocampal tissue showed that compared with non-demented controls, angiotensinogen staining was higher in Alzheimer's disease and co-localized with phosphorylated-tau. The identification of biomarker-driven endophenotypes in cognitive disorder patients further highlights the biological heterogeneity of Alzheimer's disease and the importance of tailored prevention and treatment strategies.
Collapse
Affiliation(s)
- Makrina Daniilidou
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Francesca Eroli
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Vilma Alanko
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Julen Goikolea
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Maria Latorre-Leal
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Patricia Rodriguez-Rodriguez
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | | | - Yuqin Wang
- Swansea University Medical School, Swansea SA2 8PP, UK
| | | | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 90 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 90 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 90 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 90 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N3AR, UK
- UK Dementia Research Institute at UCL, London WC1N3AR, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 90 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 90 Mölndal, Sweden
| | - Anna Rosenberg
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, FI-70029 Kuopio, Finland
| | - Nenad Bogdanovic
- Theme Inflammation and Aging, Karolinska University Hospital, 141 83 Huddinge, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 83 Huddinge, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 83 Huddinge, Sweden
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Delphine Ibghi
- Neurodegeneration Cluster, Rare and Neurologic Disease Research Sanofi R&D, F-91380 Chilly-Mazarin, France
| | - Angel Cedazo-Minguez
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Neurodegeneration Cluster, Rare and Neurologic Disease Research Sanofi R&D, F-91380 Chilly-Mazarin, France
| | - Silvia Maioli
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Anna Matton
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 83 Huddinge, Sweden
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
23
|
Li S, Roy ER, Wang Y, Watkins T, Cao W. Modeling Alzheimer's disease in primary neurons reveals DNA damage response coupled with MAPK-DLK signaling in wild-type tau-induced neurodegeneration. RESEARCH SQUARE 2023:rs.3.rs-2617457. [PMID: 36945524 PMCID: PMC10029119 DOI: 10.21203/rs.3.rs-2617457/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Background Alzheimer's disease (AD) is the most prevalent form of neurodegeneration. Despite the well-established link between tau aggregation and clinical progression, the major pathways driven by this protein to intrinsically damage neurons are incompletely understood. Methods To model AD-relevant neurodegeneration driven by tau, we overexpressed wild-type human tau in primary mouse neurons and characterized the subsequent cellular and molecular changes. RNAseq profiling and functional investigation were performed as well. A direct comparison with a mutant human tau was conducted in detail. Results We observed substantial axonal degeneration and cell death associated with wild-type tau, a process accompanied by activated caspase 3. Mechanistically, we detected deformation of the nuclear envelope and increased DNA damage response in tau-expressing neurons. Gene profiling analysis further revealed significant alterations in the mitogen-activated protein kinase (MAPK) pathway; moreover, inhibitors of dual leucine zipper kinase (DLK) and c-Jun N-terminal kinase (JNK) were effective in alleviating wild-type human tau-induced neurodegeneration. In contrast, mutant P301L human tau was less toxic to neurons, despite causing comparable DNA damage. Axonal DLK activation induced by wild-type tau potentiated the impact of DNA damage response, resulting in overt neurotoxicity. Conclusions We have established a cellular tauopathy model highly relevant to AD and identified a functional synergy between DNA damage response and the MAPK-DLK axis in the neuronal degenerative process.
Collapse
Affiliation(s)
- Sanming Li
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ethan R Roy
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanyu Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Trent Watkins
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA. Current address: Department of Neurology, University of California at San Francisco, San Francisco, CA 94158 USA
| | - Wei Cao
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
24
|
Maina MB, Al-Hilaly YK, Serpell LC. Dityrosine cross-linking and its potential roles in Alzheimer's disease. Front Neurosci 2023; 17:1132670. [PMID: 37034163 PMCID: PMC10075315 DOI: 10.3389/fnins.2023.1132670] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/01/2023] [Indexed: 04/11/2023] Open
Abstract
Oxidative stress is a significant source of damage that accumulates during aging and contributes to Alzheimer's disease (AD) pathogenesis. Oxidation of proteins can give rise to covalent links between adjacent tyrosines known as dityrosine (DiY) cross-linking, amongst other modifications, and this observation suggests that DiY could serve as a biomarker of accumulated oxidative stress over the lifespan. Many studies have focused on understanding the contribution of DiY to AD pathogenesis and have revealed that DiY crosslinks can be found in both Aβ and tau deposits - the two key proteins involved in the formation of amyloid plaques and tau tangles, respectively. However, there is no consensus yet in the field on the impact of DiY on Aβ and tau function, aggregation, and toxicity. Here we review the current understanding of the role of DiY on Aβ and tau gathered over the last 20 years since the first observation, and discuss the effect of this modification for Aβ and tau aggregation, and its potential as a biomarker for AD.
Collapse
Affiliation(s)
- Mahmoud B. Maina
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
- Biomedical Science Research and Training Centre, College of Medical Sciences, Yobe State University, Damaturu, Nigeria
| | - Youssra K. Al-Hilaly
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
- Department of Chemistry, College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Louise C. Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
- *Correspondence: Louise C. Serpell,
| |
Collapse
|
25
|
Al-Hilaly YK, Marshall KE, Lutter L, Biasetti L, Mengham K, Harrington CR, Xue WF, Wischik CM, Serpell LC. An Additive-Free Model for Tau Self-Assembly. Methods Mol Biol 2023; 2551:163-188. [PMID: 36310203 DOI: 10.1007/978-1-0716-2597-2_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Tau is a natively unfolded protein that contributes to the stability of microtubules. Under pathological conditions such as Alzheimer's disease (AD), tau protein misfolds and self-assembles to form paired helical filaments (PHFs) and straight filaments (SFs). Full-length tau protein assembles poorly and its self-assembly is enhanced with polyanions such as heparin and RNA in vitro, but a role for heparin or other polyanions in vivo remains unclear. Recently, a truncated form of tau (297-391) has been shown to self-assemble in the absence of additives which provides an alternative in vitro PHF model system. Here we describe methods to prepare in vitro PHFs and SFs from tau (297-391) named dGAE. We also discuss the range of biophysical/biochemical techniques used to monitor tau filament assembly and structure.
Collapse
Affiliation(s)
- Youssra K Al-Hilaly
- Chemistry Department, College of Science, Mustansiriyah University, Baghdad, Iraq.
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK.
| | - Karen E Marshall
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| | - Liisa Lutter
- School of Biosciences, University of Kent, Canterbury, UK
- Institute of Genomics and Proteomics, University of California, Los Angeles, CA, USA
| | - Luca Biasetti
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| | - Kurtis Mengham
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| | - Charles R Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- TauRx Therapeutics Ltd., Aberdeen, UK
| | - Wei-Feng Xue
- School of Biosciences, University of Kent, Canterbury, UK
| | - Claude M Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- TauRx Therapeutics Ltd., Aberdeen, UK
| | - Louise C Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
| |
Collapse
|
26
|
Chu S, Moujaber O, Lemay S, Stochaj U. Multiple pathways promote microtubule stabilization in senescent intestinal epithelial cells. NPJ AGING 2022; 8:16. [PMID: 36526654 PMCID: PMC9758230 DOI: 10.1038/s41514-022-00097-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Intestinal epithelial cells are critical for gastrointestinal homeostasis. However, their function declines during aging. The aging-related loss of organ performance is largely driven by the increase in senescent cells. To date, the hallmarks and molecular mechanisms related to cellular senescence are not fully understood. Microtubules control epithelial functions, and we identified microtubule stabilization as a phenotypic marker of senescent intestinal epithelial cells. The senescence inducer determined the pathway to microtubule stabilization. Specifically, enhanced microtubule stability was associated with α-tubulin hyperacetylation or increased abundance of the microtubule-binding protein tau. We show further that overexpression of MAPT, which encodes tau, augmented microtubule stability in intestinal epithelial cells. Notably, pharmacological microtubule stabilization was sufficient to induce cellular senescence. Taken together, this study provides new insights into the molecular mechanisms that control epithelial cell homeostasis. Our results support the concept that microtubule stability serves as a critical cue to trigger intestinal epithelial cell senescence.
Collapse
Affiliation(s)
- Siwei Chu
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6 Canada
| | - Ossama Moujaber
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6 Canada
| | - Serge Lemay
- grid.63984.300000 0000 9064 4811Department of Medicine, Division of Nephrology, McGill University Health Centre, Montreal, QC Canada
| | - Ursula Stochaj
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6 Canada
| |
Collapse
|
27
|
Al-Hilaly YK, Hurt C, Rickard JE, Harrington CR, Storey JMD, Wischik CM, Serpell LC, Siemer AB. Solid-state NMR of paired helical filaments formed by the core tau fragment tau(297-391). Front Neurosci 2022; 16:988074. [PMID: 36570831 PMCID: PMC9774000 DOI: 10.3389/fnins.2022.988074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Aggregation of the tau protein into fibrillar cross-β aggregates is a hallmark of Alzheimer's diseases (AD) and many other neurodegenerative tauopathies. Recently, several core structures of patient-derived tau paired helical filaments (PHFs) have been solved revealing a structural variability that often correlates with a specific tauopathy. To further characterize the dynamics of these fibril cores, to screen for strain-specific small molecules as potential biomarkers and therapeutics, and to develop strain-specific antibodies, recombinant in-vitro models of tau filaments are needed. We recently showed that a 95-residue fragment of tau (from residue 297 to 391), termed dGAE, forms filaments in vitro in the absence of polyanionic co-factors often used for in vitro aggregation of full-length tau. Tau(297-391) was identified as the proteolytic resistant core of tau PHFs and overlaps with the structures characterized by cryo-electron microscopy in ex vivo PHFs, making it a promising model for the study of AD tau filaments in vitro. In the present study, we used solid-state NMR to characterize tau(297-391) filaments and show that such filaments assembled under non-reducing conditions are more dynamic and less ordered than those made in the presence of the reducing agent DTT. We further report the resonance assignment of tau(297-391)+DTT filaments and compare it to existing core structures of tau.
Collapse
Affiliation(s)
- Youssra K. Al-Hilaly
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
- Chemistry Department, College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Connor Hurt
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Janet E. Rickard
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Charles R. Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- TauRx Therapeutics Ltd., Aberdeen, United Kingdom
| | - John M. D. Storey
- TauRx Therapeutics Ltd., Aberdeen, United Kingdom
- Department of Chemistry, University of Aberdeen, Aberdeen, United Kingdom
| | - Claude M. Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- TauRx Therapeutics Ltd., Aberdeen, United Kingdom
| | - Louise C. Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, United Kingdom
| | - Ansgar B. Siemer
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
28
|
Shapeshifting tau: from intrinsically disordered to paired-helical filaments. Essays Biochem 2022; 66:1001-1011. [PMID: 36373666 PMCID: PMC9760425 DOI: 10.1042/ebc20220150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022]
Abstract
Tau is an intrinsically disordered protein that has the ability to self-assemble to form paired helical and straight filaments in Alzheimer's disease, as well as the ability to form additional distinct tau filaments in other tauopathies. In the presence of microtubules, tau forms an elongated form associated with tubulin dimers via a series of imperfect repeats known as the microtubule binding repeats. Tau has recently been identified to have the ability to phase separate in vitro and in cells. The ability of tau to adopt a wide variety of conformations appears fundamental both to its biological function and also its association with neurodegenerative diseases. The recently highlighted involvement of low-complexity domains in liquid-liquid phase separation provides a critical link between the soluble function and the insoluble dysfunctional properties of tau.
Collapse
|
29
|
Tu J, Zhang H, Yang T, Liu Y, Kibreab S, Zhang Y, Gao L, Moses RE, O'Malley BW, Xiao J, Li X. Aging-associated REGγ proteasome decline predisposes to tauopathy. J Biol Chem 2022; 298:102571. [PMID: 36209822 PMCID: PMC9647549 DOI: 10.1016/j.jbc.2022.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/20/2022] [Accepted: 09/25/2022] [Indexed: 11/09/2022] Open
Abstract
The REGγ-20S proteasome is an ubiquitin- and ATP-independent degradation system, targeting selective substrates, possibly helping to regulate aging. The studies we report here demonstrate that aging-associated REGγ decline predisposes to decreasing tau turnover, as in a tauopathy. The REGγ proteasome promotes degradation of human and mouse tau, notably phosphorylated tau and toxic tau oligomers that shuttle between the cytoplasm and nuclei. REGγ-mediated proteasomal degradation of tau was validated in 3- to 12-month-old REGγ KO mice, REGγ KO;PS19 mice, and PS19 mice with forebrain conditional neuron-specific overexpression of REGγ (REGγ OE) and behavioral abnormalities. Coupled with tau accumulation, we found with REGγ-deficiency, neuron loss, dendrite reduction, tau filament accumulation, and microglial activation are much more prominent in the REGγ KO;PS19 than the PS19 model. Moreover, we observed that the degenerative neuronal lesions and aberrant behaviors were alleviated in REGγ OE;PS19 mice. Memory and other behavior analysis substantiate the role of REGγ in prevention of tauopathy-like symptoms. In addition, we investigated the potential mechanism underlying aging-related REGγ decline. This study provides valuable insights into the novel regulatory mechanisms and potential therapeutic targets for tau-related neurodegenerative diseases.
Collapse
|
30
|
Hedna R, Kovacic H, Pagano A, Peyrot V, Robin M, Devred F, Breuzard G. Tau Protein as Therapeutic Target for Cancer? Focus on Glioblastoma. Cancers (Basel) 2022; 14:5386. [PMID: 36358803 PMCID: PMC9653627 DOI: 10.3390/cancers14215386] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 08/27/2023] Open
Abstract
Despite being extensively studied for several decades, the microtubule-associated protein Tau has not finished revealing its secrets. For long, Tau has been known for its ability to promote microtubule assembly. A less known feature of Tau is its capability to bind to cancer-related protein kinases, suggesting a possible role of Tau in modulating microtubule-independent cellular pathways that are associated with oncogenesis. With the intention of finding new therapeutic targets for cancer, it appears essential to examine the interaction of Tau with these kinases and their consequences. This review aims at collecting the literature data supporting the relationship between Tau and cancer with a particular focus on glioblastoma tumors in which the pathological significance of Tau remains largely unexplored. We will first treat this subject from a mechanistic point of view showing the pivotal role of Tau in oncogenic processes. Then, we will discuss the involvement of Tau in dysregulating critical pathways in glioblastoma. Finally, we will outline promising strategies to target Tau protein for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Rayane Hedna
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Hervé Kovacic
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Alessandra Pagano
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Vincent Peyrot
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Maxime Robin
- Faculté de Pharmacie, Institut Méditerranéen de Biodiversité et Ecologie marine et continentale (IMBE), UMR 7263, CNRS, IRD 237, Aix-Marseille Université, 13005 Marseille, France
| | - François Devred
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Gilles Breuzard
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| |
Collapse
|
31
|
Pathological Nuclear Hallmarks in Dentate Granule Cells of Alzheimer’s Patients: A Biphasic Regulation of Neurogenesis. Int J Mol Sci 2022; 23:ijms232112873. [PMID: 36361662 PMCID: PMC9654738 DOI: 10.3390/ijms232112873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022] Open
Abstract
The dentate gyrus (DG) of the human hippocampus is a complex and dynamic structure harboring mature and immature granular neurons in diverse proliferative states. While most mammals show persistent neurogenesis through adulthood, human neurogenesis is still under debate. We found nuclear alterations in granular cells in autopsied human brains, detected by immunohistochemistry. These alterations differ from those reported in pyramidal neurons of the hippocampal circuit. Aging and early AD chromatin were clearly differentiated by the increased epigenetic markers H3K9me3 (heterochromatin suppressive mark) and H3K4me3 (transcriptional euchromatin mark). At early AD stages, lamin B2 was redistributed to the nucleoplasm, indicating cell-cycle reactivation, probably induced by hippocampal nuclear pathology. At intermediate and late AD stages, higher lamin B2 immunopositivity in the perinucleus suggests fewer immature neurons, less neurogenesis, and fewer adaptation resources to environmental factors. In addition, senile samples showed increased nuclear Tau interacting with aged chromatin, likely favoring DNA repair and maintaining genomic stability. However, at late AD stages, the progressive disappearance of phosphorylated Tau forms in the nucleus, increased chromatin disorganization, and increased nuclear autophagy support a model of biphasic neurogenesis in AD. Therefore, designing therapies to alleviate the neuronal nuclear pathology might be the only pathway to a true rejuvenation of brain circuits.
Collapse
|
32
|
Barbolina MV. Targeting Microtubule-Associated Protein Tau in Chemotherapy-Resistant Models of High-Grade Serous Ovarian Carcinoma. Cancers (Basel) 2022; 14:4535. [PMID: 36139693 PMCID: PMC9496900 DOI: 10.3390/cancers14184535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Relapsed, recurrent, chemotherapy-resistant high-grade serous ovarian carcinoma is the deadliest stage of this disease. Expression of microtubule-associated protein tau (tau) has been linked to resistance to paclitaxel treatment. Here, I used models of platinum-resistant and created models of platinum/paclitaxel-resistant high-grade serous ovarian carcinoma to examine the impact of reducing tau expression on cell survival and tumor burden in cell culture and xenograft and syngeneic models of the disease. Tau was overexpressed in platinum/paclitaxel-resistant models; expression of phosphoSer396 and phosphoThr181 species was also found. A treatment with leucomethylene blue reduced the levels of tau in treated cells, was cytotoxic in cell cultures, and efficiently reduced the tumor burden in xenograft models. Furthermore, a combination of leucomethylene blue and paclitaxel synergized in eliminating cancer cells in cell culture and xenograft models. These findings underscore the feasibility of targeting tau as a treatment option in terminal-stage high-grade serous ovarian cancer.
Collapse
Affiliation(s)
- Maria V Barbolina
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60091, USA
| |
Collapse
|
33
|
Fourest-Lieuvin A, Vinit A, Blot B, Perrot A, Denarier E, Saudou F, Arnal I. Controlled Tau Cleavage in Cells Reveals Abnormal Localizations of Tau Fragments. Neuroscience 2022; 518:162-177. [PMID: 35995336 DOI: 10.1016/j.neuroscience.2022.08.016] [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: 12/16/2021] [Revised: 06/08/2022] [Accepted: 08/16/2022] [Indexed: 11/15/2022]
Abstract
In several forms of dementia, such as Alzheimer's disease, the cytoskeleton-associated protein tau undergoes proteolysis, giving rise to fragments that have a toxic impact on neuronal homeostasis. How these fragments interact with cellular structures, in particular with the cytoskeleton, is currently incompletely understood. Here, we developed a method, derived from a Tobacco Etch Virus (TEV) protease system, to induce controlled cleavage of tau at specific sites. Five tau proteins containing specific TEV recognition sites corresponding to pathological proteolytic sites were engineered, and tagged with GFP at one end and mCherry at the other. Following controlled cleavage to produce GFP-N-terminal and C-terminal-mCherry fragments, we followed the fate of tau fragments in cells. Our results showed that whole engineered tau proteins associate with the cytoskeleton similarly to the non-modified tau, whereas tau fragments adopted different localizations with respect to the actin and microtubule cytoskeletons. These distinct localizations were confirmed by expressing each separate fragment in cells. Some cleavages - in particular cleavages at amino-acid positions 124 or 256 - displayed a certain level of cellular toxicity, with an unusual relocalization of the N-terminal fragments to the nucleus. Based on the data presented here, inducible cleavage of tau by the TEV protease appears to be a valuable tool to reproduce tau fragmentation in cells and study the resulting consequences on cell physiology.
Collapse
Affiliation(s)
- Anne Fourest-Lieuvin
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| | - Angélique Vinit
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Béatrice Blot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Anthime Perrot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eric Denarier
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frédéric Saudou
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Isabelle Arnal
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
34
|
Jackson NA, Guerrero-Muñoz MJ, Castillo-Carranza DL. The prion-like transmission of tau oligomers via exosomes. Front Aging Neurosci 2022; 14:974414. [PMID: 36062141 PMCID: PMC9434014 DOI: 10.3389/fnagi.2022.974414] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
The conversion and transmission of misfolded proteins established the basis for the prion concept. Neurodegenerative diseases are considered “prion-like” disorders that lack infectivity. Among them, tauopathies are characterized by the conversion of native tau protein into an abnormally folded aggregate. During the progression of the disease, misfolded tau polymerizes into oligomers and intracellular neurofibrillary tangles (NFTs). While the toxicity of NFTs is an ongoing debate, the contribution of tau oligomers to early onset neurodegenerative pathogenesis is accepted. Tau oligomers are readily transferred from neuron to neuron propagating through the brain inducing neurodegeneration. Recently, transmission of tau oligomers via exosomes is now proposed. There is still too much to uncover about tau misfolding and propagation. Here we summarize novel findings of tau oligomers transmission and propagation via exosomes.
Collapse
Affiliation(s)
- Noel A. Jackson
- School of Public Health, Harvard University, Boston, MA, United States
| | | | - Diana L. Castillo-Carranza
- School of Medicine, University of Monterrey, San Pedro Garza García, Mexico
- *Correspondence: Diana L. Castillo-Carranza,
| |
Collapse
|
35
|
Maina MB, Al-Hilaly YK, Oakley S, Burra G, Khanom T, Biasetti L, Mengham K, Marshall K, Harrington CR, Wischik CM, Serpell LC. Dityrosine Cross-links are Present in Alzheimer's Disease-derived Tau Oligomers and Paired Helical Filaments (PHF) which Promotes the Stability of the PHF-core Tau (297-391) In Vitro. J Mol Biol 2022; 434:167785. [PMID: 35961386 DOI: 10.1016/j.jmb.2022.167785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
Abstract
A characteristic hallmark of Alzheimer's Disease (AD) is the pathological aggregation and deposition of tau into paired helical filaments (PHF) in neurofibrillary tangles (NFTs). Oxidative stress is an early event during AD pathogenesis and is associated with tau-mediated AD pathology. Oxidative environments can result in the formation of covalent dityrosine crosslinks that can increase protein stability and insolubility. Dityrosine cross-linking has been shown in Aβ plaques in AD and α-synuclein aggregates in Lewy bodies in ex vivo tissue sections, and this modification may increase the insolubility of these aggregates and their resistance to degradation. Using the PHF-core tau fragment (residues 297 - 391) as a model, we have previously demonstrated that dityrosine formation traps tau assemblies to reduce further elongation. However, it is unknown whether dityrosine crosslinks are found in tau deposits in vivo in AD and its relevance to disease mechanism is unclear. Here, using transmission electron microscope (TEM) double immunogold-labelling, we reveal that neurofibrillary NFTs in AD are heavily decorated with dityrosine crosslinks alongside tau. Single immunogold-labelling TEM and fluorescence spectroscopy revealed the presence of dityrosine on AD brain-derived tau oligomers and fibrils. Using the tau (297-391) PHF-core fragment as a model, we further showed that prefibrillar tau species are more amenable to dityrosine crosslinking than tau fibrils. Dityrosine formation results in heat and SDS stability of oxidised prefibrillar and fibrillar tau assemblies. This finding has implications for understanding the mechanism governing the insolubility and toxicity of tau assemblies in vivo.
Collapse
Affiliation(s)
- Mahmoud B Maina
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK; Biomedical Science Research and Training Centre, Yobe State University, Nigeria. https://twitter.com/mahmoudbukar
| | - Youssra K Al-Hilaly
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK; Chemistry Department, College of Sciences, Mustansiriyah University, Baghdad, Iraq
| | - Sebastian Oakley
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Gunasekhar Burra
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK; Analytical Development Biologics, Biopharmaceutical Development, Syngene International Limited, Biocon Park, Bommasandra Jigani Link Road, Bangalore 560009, India
| | - Tahmida Khanom
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Luca Biasetti
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Kurtis Mengham
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Karen Marshall
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK
| | - Charles R Harrington
- Institute of Medical Sciences, University of Aberdeen, UK; TauRx Therapeutics Ltd, Aberdeen, UK
| | - Claude M Wischik
- Institute of Medical Sciences, University of Aberdeen, UK; TauRx Therapeutics Ltd, Aberdeen, UK
| | - Louise C Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex UK.
| |
Collapse
|
36
|
Singh S, Yang F, Sivils A, Cegielski V, Chu XP. Amylin and Secretases in the Pathology and Treatment of Alzheimer's Disease. Biomolecules 2022; 12:996. [PMID: 35883551 PMCID: PMC9312829 DOI: 10.3390/biom12070996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease remains a prevailing neurodegenerative condition which has an array physical, emotional, and financial consequences to patients and society. In the past decade, there has been a greater degree of investigation on therapeutic small peptides. This group of biomolecules have a profile of fundamentally sound characteristics which make them an intriguing area for drug development. Among these biomolecules, there are four modulatory mechanisms of interest in this review: alpha-, beta-, gamma-secretases, and amylin. These protease-based biomolecules all have a contributory role in the amyloid cascade hypothesis. Moreover, the involvement of various biochemical pathways intertwines these peptides to have shared regulators (i.e., retinoids). Further clinical and translational investigation must occur to gain a greater understanding of its potential application in patient care. The aim of this narrative review is to evaluate the contemporary literature on these protease biomolecule modulators and determine its utility in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Xiang-Ping Chu
- Department of Biomedical Sciences, School of Medicine, University of Missouri, Kansas City, MO 64108, USA; (S.S.); (F.Y.); (A.S.); (V.C.)
| |
Collapse
|
37
|
Tau as a Biomarker of Neurodegeneration. Int J Mol Sci 2022; 23:ijms23137307. [PMID: 35806324 PMCID: PMC9266883 DOI: 10.3390/ijms23137307] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022] Open
Abstract
Less than 50 years since tau was first isolated from a porcine brain, its detection in femtolitre concentrations in biological fluids is revolutionizing the diagnosis of neurodegenerative diseases. This review highlights the molecular and technological advances that have catapulted tau from obscurity to the forefront of biomarker diagnostics. Comprehensive updates are provided describing the burgeoning clinical applications of tau as a biomarker of neurodegeneration. For the clinician, tau not only enhances diagnostic accuracy, but holds promise as a predictor of clinical progression, phenotype, and response to drug therapy. For patients living with neurodegenerative disorders, characterization of tau dysregulation could provide much-needed clarity to a notoriously murky diagnostic landscape.
Collapse
|
38
|
Nelson RS, Dammer EB, Santiago JV, Seyfried NT, Rangaraju S. Brain Cell Type-Specific Nuclear Proteomics Is Imperative to Resolve Neurodegenerative Disease Mechanisms. Front Neurosci 2022; 16:902146. [PMID: 35784845 PMCID: PMC9243337 DOI: 10.3389/fnins.2022.902146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/30/2022] [Indexed: 01/19/2023] Open
Abstract
Neurodegenerative diseases (NDs) involve complex cellular mechanisms that are incompletely understood. Emerging findings have revealed that disruption of nuclear processes play key roles in ND pathogenesis. The nucleus is a nexus for gene regulation and cellular processes that together, may underlie pathomechanisms of NDs. Furthermore, many genetic risk factors for NDs encode proteins that are either present in the nucleus or are involved in nuclear processes (for example, RNA binding proteins, epigenetic regulators, or nuclear-cytoplasmic transport proteins). While recent advances in nuclear transcriptomics have been significant, studies of the nuclear proteome in brain have been relatively limited. We propose that a comprehensive analysis of nuclear proteomic alterations of various brain cell types in NDs may provide novel biological and therapeutic insights. This may be feasible because emerging technical advances allow isolation and investigation of intact nuclei from post-mortem frozen human brain tissue with cell type-specific and single-cell resolution. Accordingly, nuclei of various brain cell types harbor unique protein markers which can be used to isolate cell-type specific nuclei followed by down-stream proteomics by mass spectrometry. Here we review the literature providing a rationale for investigating proteomic changes occurring in nuclei in NDs and then highlight the potential for brain cell type-specific nuclear proteomics to enhance our understanding of distinct cellular mechanisms that drive ND pathogenesis.
Collapse
Affiliation(s)
- Ruth S. Nelson
- Department of Neurology, Emory University, Atlanta, GA, United States
| | - Eric B. Dammer
- Department of Biochemistry, Emory University, Atlanta, GA, United States
| | | | | | - Srikant Rangaraju
- Department of Neurology, Emory University, Atlanta, GA, United States,*Correspondence: Srikant Rangaraju
| |
Collapse
|
39
|
Latimer CS, Stair JG, Hincks JC, Currey HN, Bird TD, Keene CD, Kraemer BC, Liachko NF. TDP-43 promotes tau accumulation and selective neurotoxicity in bigenic Caenorhabditis elegans. Dis Model Mech 2022; 15:275149. [PMID: 35178571 PMCID: PMC9066518 DOI: 10.1242/dmm.049323] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022] Open
Abstract
Although amyloid β (Aβ) and tau aggregates define the neuropathology of Alzheimer's disease (AD), TDP-43 has recently emerged as a co-morbid pathology in more than half of patients with AD. Individuals with concomitant Aβ, tau and TDP-43 pathology experience accelerated cognitive decline and worsened brain atrophy, but the molecular mechanisms of TDP-43 neurotoxicity in AD are unknown. Synergistic interactions among Aβ, tau and TDP-43 may be responsible for worsened disease outcomes. To study the biology underlying this process, we have developed new models of protein co-morbidity using the simple animal Caenorhabditis elegans. We demonstrate that TDP-43 specifically enhances tau but not Aβ neurotoxicity, resulting in neuronal dysfunction, pathological tau accumulation and selective neurodegeneration. Furthermore, we find that synergism between tau and TDP-43 is rescued by loss-of-function of the robust tau modifier sut-2. Our results implicate enhanced tau neurotoxicity as the primary driver underlying worsened clinical and neuropathological phenotypes in AD with TDP-43 pathology, and identify cell-type specific sensitivities to co-morbid tau and TDP-43. Determining the relationship between co-morbid TDP-43 and tau is crucial to understand, and ultimately treat, mixed pathology AD.
Collapse
Affiliation(s)
- Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Jade G. Stair
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Joshua C. Hincks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Heather N. Currey
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Thomas D. Bird
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Department of Neurology, University of Washington, Seattle, WA 98104, USA,Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Brian C. Kraemer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA,Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Nicole F. Liachko
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA,Author for correspondence ()
| |
Collapse
|
40
|
Tsolaki E, Csincsik L, Xue J, Lengyel I, Bertazzo S. Nuclear and cellular, micro and nano calcification in Alzheimer's disease patients and correlation to phosphorylated Tau. Acta Biomater 2022; 143:138-144. [PMID: 35259518 DOI: 10.1016/j.actbio.2022.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 12/26/2022]
Abstract
Brain calcification (calcium phosphate mineral formation) has been reported in the past 100 years in the brains of Alzheimer's disease (AD) patients. However, the association between calcification and AD, the triggers for calcification, and its role within the disease are not clear. On the other hand, hyperphosphorylated Tau protein (pTau) tangles have been widely studied and recognized as an essential factor in developing AD. In this work, calcification in the brains of AD patients is characterized by advanced electron microscopy and fluorescence microscopy. Results are then compared to samples from cognitively healthy, age-matched donors, and the colocalization of calcification and pTau is investigated. Here, we show that AD patients' brains present microcalcification associated with the neural cell nuclei and cell projections, and that these are strongly related to the presence of pTau. The link between microcalcification and pTau suggests a potential mechanism of brain cell damage. Together with the formation of amyloid plaques and neurofibrillary tangles, microcalcification in neuronal cells adds to a better understanding of the pathology of AD. Finally, the presence of microcalcification in the neuronal cells of AD patients may assist in AD diagnosis, and may open avenues for developing intervention strategies based on inhibition of calcification. STATEMENT OF SIGNIFICANCE: Brain calcification has been reported in the past 100 years in the brains of Alzheimer's disease (AD) patients. However, the association between calcification and AD is not clear. Hyperphosphorylated Tau protein (pTau) has been studied and recognized as a key factor in developing AD. We show here that AD patients' brains present microcalcification associated with the neuronal cell nuclei and cell projections, and that these are related to the presence of pTau. The study of calcification in brain cells can contribute to a better understanding of the biochemical mechanisms associated with AD and might also reveal that calcification is part of the full disease mechanism. Moreover, this work opens the possibility for using calcification as a biomarker to identify AD.
Collapse
|
41
|
Ruiz-Gabarre D, Carnero-Espejo A, Ávila J, García-Escudero V. What's in a Gene? The Outstanding Diversity of MAPT. Cells 2022; 11:840. [PMID: 35269461 PMCID: PMC8909800 DOI: 10.3390/cells11050840] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 02/04/2023] Open
Abstract
Tau protein is a microtubule-associated protein encoded by the MAPT gene that carries out a myriad of physiological functions and has been linked to certain pathologies collectively termed tauopathies, including Alzheimer's disease, frontotemporal dementia, Huntington's disease, progressive supranuclear palsy, etc. Alternative splicing is a physiological process by which cells generate several transcripts from one single gene and may in turn give rise to different proteins from the same gene. MAPT transcripts have been proven to be subjected to alternative splicing, generating six main isoforms in the central nervous system. Research throughout the years has demonstrated that the splicing landscape of the MAPT gene is far more complex than that, including at least exon skipping events, the use of 3' and 5' alternative splice sites and, as has been recently discovered, also intron retention. In addition, MAPT alternative splicing has been showed to be regulated spatially and developmentally, further evidencing the complexity of the gene's splicing regulation. It is unclear what would drive the need for the existence of so many isoforms encoded by the same gene, but a wide range of functions have been ascribed to these Tau isoforms, both in physiology and pathology. In this review we offer a comprehensive up-to-date exploration of the mechanisms leading to the outstanding diversity of isoforms expressed from the MAPT gene and the functions in which such isoforms are involved, including their potential role in the onset and development of tauopathies such as Alzheimer's disease.
Collapse
Affiliation(s)
- Daniel Ruiz-Gabarre
- Anatomy, Histology and Neuroscience Department, School of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain; (D.R.-G.); (A.C.-E.)
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), 28049 Madrid, Spain
- Graduate Program in Neuroscience, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Almudena Carnero-Espejo
- Anatomy, Histology and Neuroscience Department, School of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain; (D.R.-G.); (A.C.-E.)
- Graduate Program in Neuroscience, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Jesús Ávila
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), 28049 Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Vega García-Escudero
- Anatomy, Histology and Neuroscience Department, School of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain; (D.R.-G.); (A.C.-E.)
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), 28049 Madrid, Spain
- Graduate Program in Neuroscience, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| |
Collapse
|
42
|
Sauna-like conditions or menthol treatment reduce tau phosphorylation through mild hyperthermia. Neurobiol Aging 2022; 113:118-130. [DOI: 10.1016/j.neurobiolaging.2022.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 02/02/2022] [Accepted: 02/20/2022] [Indexed: 02/08/2023]
|
43
|
Mold MJ, Exley C. Aluminium co-localises with Biondi ring tangles in Parkinson's disease and epilepsy. Sci Rep 2022; 12:1465. [PMID: 35087154 PMCID: PMC8795119 DOI: 10.1038/s41598-022-05627-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/12/2022] [Indexed: 12/20/2022] Open
Abstract
Aluminium is known to accumulate in neuropathological hallmarks. However, such has only tentatively been suggested in Biondi ring tangles. Owing to their intracellular and filamentous structure rich in β-pleated sheets, Biondi ring tangles might attract the adventitious binding of aluminium in regions of the blood-cerebrospinal fluid barrier. The study's objective was to establish whether aluminium co-localises with Biondi ring tangles in the brains of Parkinson's disease donors versus a donor that went on to develop late-onset epilepsy. Herein, we have performed immunohistochemistry for phosphorylated tau, complemented with aluminium-specific fluorescence microscopy in the choroid plexus of Parkinson's disease donors and in a donor that developed late-onset epilepsy. Aluminium co-localises with lipid-rich Biondi ring tangles in the choroid plexus. While Biondi ring tangles are not composed of phosphorylated tau, the latter is identified in nuclei of choroidal cells where aluminium and Biondi ring tangles are co-located. Although Biondi ring tangles are considered artefacts in imaging studies using positron emission tomography, their ability to bind aluminium and then release it upon their subsequent rupture and escape from choroidal cells may allow for a mechanism that may propagate for aluminium toxicity in vivo.
Collapse
Affiliation(s)
- Matthew John Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK
| |
Collapse
|
44
|
Tau mRNA Metabolism in Neurodegenerative Diseases: A Tangle Journey. Biomedicines 2022; 10:biomedicines10020241. [PMID: 35203451 PMCID: PMC8869323 DOI: 10.3390/biomedicines10020241] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 12/07/2022] Open
Abstract
Tau proteins are known to be mainly involved in regulation of microtubule dynamics. Besides this function, which is critical for axonal transport and signal transduction, tau proteins also have other roles in neurons. Moreover, tau proteins are turned into aggregates and consequently trigger many neurodegenerative diseases termed tauopathies, of which Alzheimer’s disease (AD) is the figurehead. Such pathological aggregation processes are critical for the onset of these diseases. Among the various causes of tau protein pathogenicity, abnormal tau mRNA metabolism, expression and dysregulation of tau post-translational modifications are critical steps. Moreover, the relevance of tau function to general mRNA metabolism has been highlighted recently in tauopathies. In this review, we mainly focus on how mRNA metabolism impacts the onset and development of tauopathies. Thus, we intend to portray how mRNA metabolism of, or mediated by, tau is associated with neurodegenerative diseases.
Collapse
|
45
|
Rico T, Gilles M, Chauderlier A, Comptdaer T, Magnez R, Chwastyniak M, Drobecq H, Pinet F, Thuru X, Buée L, Galas MC, Lefebvre B. Tau Stabilizes Chromatin Compaction. Front Cell Dev Biol 2021; 9:740550. [PMID: 34722523 PMCID: PMC8551707 DOI: 10.3389/fcell.2021.740550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
An extensive body of literature suggested a possible role of the microtubule-associated protein Tau in chromatin functions and/or organization in neuronal, non-neuronal, and cancer cells. How Tau functions in these processes remains elusive. Here we report that Tau expression in breast cancer cell lines causes resistance to the anti-cancer effects of histone deacetylase inhibitors, by preventing histone deacetylase inhibitor-inducible gene expression and remodeling of chromatin structure. We identify Tau as a protein recognizing and binding to core histone when H3 and H4 are devoid of any post-translational modifications or acetylated H4 that increases the Tau's affinity. Consistent with chromatin structure alterations in neurons found in frontotemporal lobar degeneration, Tau mutations did not prevent histone deacetylase-inhibitor-induced higher chromatin structure remodeling by suppressing Tau binding to histones. In addition, we demonstrate that the interaction between Tau and histones prevents further histone H3 post-translational modifications induced by histone deacetylase-inhibitor treatment by maintaining a more compact chromatin structure. Altogether, these results highlight a new cellular role for Tau as a chromatin reader, which opens new therapeutic avenues to exploit Tau biology in neuronal and cancer cells.
Collapse
Affiliation(s)
- Thomas Rico
- Univ. Lille, INSERM, CHU-Lille, Lille Neuroscience and Cognition, UMR-S1172, Alzheimer and Tauopathies, Lille, France
| | - Melissa Gilles
- Univ. Lille, INSERM, CHU-Lille, Lille Neuroscience and Cognition, UMR-S1172, Alzheimer and Tauopathies, Lille, France
| | - Alban Chauderlier
- Univ. Lille, INSERM, CHU-Lille, Lille Neuroscience and Cognition, UMR-S1172, Alzheimer and Tauopathies, Lille, France
| | - Thomas Comptdaer
- Univ. Lille, INSERM, CHU-Lille, Lille Neuroscience and Cognition, UMR-S1172, Alzheimer and Tauopathies, Lille, France
| | - Romain Magnez
- Univ. Lille, CNRS, INSERM, CHU Lille, UMR 9020, UMR 1277, Canther, Platform of Integrative Chemical Biology, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Maggy Chwastyniak
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées Au Vieillissement, Lille, France
| | - Herve Drobecq
- Univ. Lille, CNRS UMR 9017, INSERM U1019, CHRU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
| | - Florence Pinet
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées Au Vieillissement, Lille, France
| | - Xavier Thuru
- Univ. Lille, CNRS, INSERM, CHU Lille, UMR 9020, UMR 1277, Canther, Platform of Integrative Chemical Biology, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Luc Buée
- Univ. Lille, INSERM, CHU-Lille, Lille Neuroscience and Cognition, UMR-S1172, Alzheimer and Tauopathies, Lille, France
| | - Marie-Christine Galas
- Univ. Lille, INSERM, CHU-Lille, Lille Neuroscience and Cognition, UMR-S1172, Alzheimer and Tauopathies, Lille, France
| | - Bruno Lefebvre
- Univ. Lille, INSERM, CHU-Lille, Lille Neuroscience and Cognition, UMR-S1172, Alzheimer and Tauopathies, Lille, France
| |
Collapse
|
46
|
Carroll T, Guha S, Nehrke K, Johnson GVW. Tau Post-Translational Modifications: Potentiators of Selective Vulnerability in Sporadic Alzheimer's Disease. BIOLOGY 2021; 10:1047. [PMID: 34681146 PMCID: PMC8533264 DOI: 10.3390/biology10101047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022]
Abstract
Sporadic Alzheimer's Disease (AD) is the most common form of dementia, and its severity is characterized by the progressive formation of tau neurofibrillary tangles along a well-described path through the brain. This spatial progression provides the basis for Braak staging of the pathological progression for AD. Tau protein is a necessary component of AD pathology, and recent studies have found that soluble tau species with selectively, but not extensively, modified epitopes accumulate along the path of disease progression before AD-associated insoluble aggregates form. As such, modified tau may represent a key cellular stressing agent that potentiates selective vulnerability in susceptible neurons during AD progression. Specifically, studies have found that tau phosphorylated at sites such as T181, T231, and S396 may initiate early pathological changes in tau by disrupting proper tau localization, initiating tau oligomerization, and facilitating tau accumulation and extracellular export. Thus, this review elucidates potential mechanisms through which tau post-translational modifications (PTMs) may simultaneously serve as key modulators of the spatial progression observed in AD development and as key instigators of early pathology related to neurodegeneration-relevant cellular dysfunctions.
Collapse
Affiliation(s)
- Trae Carroll
- Department of Pathology, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Sanjib Guha
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| |
Collapse
|
47
|
Sexton C, Snyder H, Beher D, Boxer AL, Brannelly P, Brion JP, Buée L, Cacace AM, Chételat G, Citron M, DeVos SL, Diaz K, Feldman HH, Frost B, Goate AM, Gold M, Hyman B, Johnson K, Karch CM, Kerwin DR, Koroshetz WJ, Litvan I, Morris HR, Mummery CJ, Mutamba J, Patterson MC, Quiroz YT, Rabinovici GD, Rommel A, Shulman MB, Toledo-Sherman LM, Weninger S, Wildsmith KR, Worley SL, Carrillo MC. Current directions in tau research: Highlights from Tau 2020. Alzheimers Dement 2021; 18:988-1007. [PMID: 34581500 DOI: 10.1002/alz.12452] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 11/07/2022]
Abstract
Studies supporting a strong association between tau deposition and neuronal loss, neurodegeneration, and cognitive decline have heightened the allure of tau and tau-related mechanisms as therapeutic targets. In February 2020, leading tau experts from around the world convened for the first-ever Tau2020 Global Conference in Washington, DC, co-organized and cosponsored by the Rainwater Charitable Foundation, the Alzheimer's Association, and CurePSP. Representing academia, industry, government, and the philanthropic sector, presenters and attendees discussed recent advances and current directions in tau research. The meeting provided a unique opportunity to move tau research forward by fostering global partnerships among academia, industry, and other stakeholders and by providing support for new drug discovery programs, groundbreaking research, and emerging tau researchers. The meeting also provided an opportunity for experts to present critical research-advancing tools and insights that are now rapidly accelerating the pace of tau research.
Collapse
Affiliation(s)
| | | | | | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Pat Brannelly
- Alzheimer's Disease Data Initiative, Kirkland, WI, USA
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Luc Buée
- Univ Lille, Inserm, CHU-Lille, Lille Neuroscience and Cognition, Place de Verdun, Lille, France
| | | | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, France
| | - Martin Citron
- Neuroscience TA, Braine l'Alleud, UCB Biopharma, Brussels, Belgium
| | - Sarah L DeVos
- Translational Sciences, Denali Therapeutics, San Francisco, California, USA
| | | | - Howard H Feldman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Bess Frost
- Sam & Ann Barshop Institute for Longevity and Aging Studies, Glenn Biggs Institute for Alzheimer's & Neurodegenerative Disorders, Department of Cell Systems & Anatomy, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Neuroscience, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Gold
- AbbVie, Neurosciences Development, North Chicago, Illinois, USA
| | - Bradley Hyman
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Johnson
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Diana R Kerwin
- Kerwin Medical Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Walter J Koroshetz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, San Diego, California, USA
| | - Huw R Morris
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | - Catherine J Mummery
- Dementia Research Centre, National Hospital for Neurology and Neurosurgery, University College London, London, UK
| | | | - Marc C Patterson
- Departments of Neurology, Pediatrics and Medical Genetics, Mayo Clinic, Rochester, Minnesota, USA
| | - Yakeel T Quiroz
- Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Departments of Neurology, Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Amy Rommel
- Tau Consortium, Rainwater Charitable Foundation, Fort Worth, Texas, USA
| | - Melanie B Shulman
- Neurodegeneration Development Unit, Biogen, Boston, Massachusetts, USA
| | | | | | - Kristin R Wildsmith
- Department of Biomarker Development, Genentech, South San Francisco, California, USA
| | - Susan L Worley
- Independent science writer, Bryn Mawr, Pennsylvania, USA
| | | |
Collapse
|
48
|
Kang SG, Han ZZ, Daude N, McNamara E, Wohlgemuth S, Molina-Porcel L, Safar JG, Mok SA, Westaway D. Pathologic tau conformer ensembles induce dynamic, liquid-liquid phase separation events at the nuclear envelope. BMC Biol 2021; 19:199. [PMID: 34503506 PMCID: PMC8428099 DOI: 10.1186/s12915-021-01132-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
Background The microtubule-associated protein tau forms aggregates in different neurodegenerative diseases called tauopathies. Prior work has shown that a single P301L mutation in tau gene, MAPT, can promote alternative tau folding pathways that correlate with divergent clinical diagnoses. Using progressive chemical denaturation, some tau preparations from the brain featured complex transitions starting at low concentrations of guanidine hydrochloride (GdnHCl) denaturant, indicating an ensemble of differently folded tau species called conformers. On the other hand, brain samples with abundant, tangle-like pathology had simple GdnHCl unfolding profile resembling the profile of fibrillized recombinant tau and suggesting a unitary conformer composition. In studies here we sought to understand tau conformer progression and potential relationships with condensed liquid states, as well as associated perturbations in cell biological processes. Results As starting material, we used brain samples from P301L transgenic mice containing tau conformer ensembles that unfolded at low GdnHCl concentrations and with signatures resembling brain material from P301L subjects presenting with language or memory problems. We seeded reporter cells expressing a soluble form of 4 microtubule-binding repeat tau fused to GFP or YFP reporter moieties, resulting in redistribution of dispersed fluorescence signals into focal assemblies that could fuse together and move within processes between adjacent cells. Nuclear envelope fluorescent tau signals and small fluorescent inclusions behaved as a demixed liquid phase, indicative of liquid-liquid phase separation (LLPS); these droplets exhibited spherical morphology, fusion events and could recover from photobleaching. Moreover, juxtanuclear tau assemblies were associated with disrupted nuclear transport and reduced cell viability in a stable cell line. Staining for thioflavin S (ThS) became more prevalent as tau-derived inclusions attained cross-sectional area greater than 3 μm2, indicating (i) a bipartite composition, (ii) in vivo progression of tau conformers, and (iii) that a mass threshold applying to demixed condensates may drive liquid-solid transitions. Conclusions Tau conformer ensembles characterized by denaturation at low GdnHCl concentration templated the production of condensed droplets in living cells. These species exhibit dynamic changes and develop in vivo, and the larger ThS-positive assemblies may represent a waystation to arrive at intracellular fibrillar tau inclusions seen in end-stage genetic tauopathies. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01132-y.
Collapse
Affiliation(s)
- Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada
| | - Zhuang Zhuang Han
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Nathalie Daude
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada
| | - Emily McNamara
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Serene Wohlgemuth
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada
| | | | - Jiri G Safar
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sue-Ann Mok
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada. .,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada. .,Division of Neurology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
49
|
D’Andrea L, Stringhi R, Di Luca M, Marcello E. Looking at Alzheimer's Disease Pathogenesis from the Nuclear Side. Biomolecules 2021; 11:biom11091261. [PMID: 34572474 PMCID: PMC8467578 DOI: 10.3390/biom11091261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 01/22/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder representing the most common form of dementia. It is biologically characterized by the deposition of extracellular amyloid-β (Aβ) senile plaques and intracellular neurofibrillary tangles, constituted by hyperphosphorylated tau protein. The key protein in AD pathogenesis is the amyloid precursor protein (APP), which is cleaved by secretases to produce several metabolites, including Aβ and APP intracellular domain (AICD). The greatest genetic risk factor associated with AD is represented by the Apolipoprotein E ε4 (APOE ε4) allele. Importantly, all of the above-mentioned molecules that are strictly related to AD pathogenesis have also been described as playing roles in the cell nucleus. Accordingly, evidence suggests that nuclear functions are compromised in AD. Furthermore, modulation of transcription maintains cellular homeostasis, and alterations in transcriptomic profiles have been found in neurodegenerative diseases. This report reviews recent advancements in the AD players-mediated gene expression. Aβ, tau, AICD, and APOE ε4 localize in the nucleus and regulate the transcription of several genes, part of which is involved in AD pathogenesis, thus suggesting that targeting nuclear functions might provide new therapeutic tools for the disease.
Collapse
|
50
|
Colussi C, Grassi C. Epigenetic regulation of neural stem cells: The emerging role of nucleoporins. STEM CELLS (DAYTON, OHIO) 2021; 39:1601-1614. [PMID: 34399020 PMCID: PMC9290943 DOI: 10.1002/stem.3444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/28/2021] [Indexed: 11/06/2022]
Abstract
Nucleoporins (Nups) are components of the nuclear pore complex that, besides regulating nucleus-cytoplasmic transport, emerged as a hub for chromatin interaction and gene expression modulation. Specifically, Nups act in a dynamic manner both at specific gene level and in the topological organization of chromatin domains. As such, they play a fundamental role during development and determination of stemness/differentiation balance in stem cells. An increasing number of reports indicate the implication of Nups in many central nervous system functions with great impact on neurogenesis, neurophysiology, and neurological disorders. Nevertheless, the role of Nup-mediated epigenetic regulation in embryonic and adult neural stem cells (NSCs) is a field largely unexplored and the comprehension of their mechanisms of action is only beginning to be unveiled. After a brief overview of epigenetic mechanisms, we will present and discuss the emerging role of Nups as new effectors of neuroepigenetics and as dynamic platform for chromatin function with specific reference to the biology of NSCs.
Collapse
Affiliation(s)
- Claudia Colussi
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI)-CNR, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|