1
|
Whitney K, Song WM, Sharma A, Dangoor DK, Farrell K, Krassner MM, Ressler HW, Christie TD, Kandoi S, Walker RH, Nirenberg MJ, Frucht SJ, Riboldi GM, Zhang B, Pereira AC, Crary JF. Single-cell transcriptomic and neuropathologic analysis reveals dysregulation of the integrated stress response in progressive supranuclear palsy. Acta Neuropathol 2024; 148:80. [PMID: 39648200 PMCID: PMC11625691 DOI: 10.1007/s00401-024-02823-w] [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: 10/27/2023] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 12/10/2024]
Abstract
Progressive supranuclear palsy (PSP) is a sporadic neurodegenerative tauopathy variably affecting brainstem and cortical structures, and characterized by tau inclusions in neurons and glia. The precise mechanism whereby these protein aggregates lead to cell death remains unclear. To investigate the contribution of these different cellular abnormalities to PSP pathogenesis, we performed single-nucleus RNA sequencing (snRNA-seq) and analyzed 50,708 high quality nuclei targeting the diencephalon, including the subthalamic nucleus and adjacent structures, from human post-mortem PSP brains with varying degrees of pathology compared to controls. Cell-type-specific differential expression and pathway analysis identified both common and discrete changes in numerous pathways previously implicated in PSP and other neurodegenerative disorders. This included EIF2 signaling, an adaptive pathway activated in response to diverse stressors, which was activated in multiple vulnerable cell types and validated in independent snRNA-seq and bulk RNA-seq datasets. Using immunohistochemistry, we found that activated eIF2α was positively correlated with tau pathology burden in vulnerable brain regions. Multiplex immunofluorescence localized activated eIF2α positivity to hyperphosphorylated tau (p-tau) positive neurons and ALDH1L1-positive astrocytes, supporting the increased transcriptomic EIF2 activation observed in these vulnerable cell types. In conclusion, these data provide insights into cell-type-specific pathological changes in PSP and support the hypothesis that failure of adaptive stress pathways play a mechanistic role in the pathogenesis and progression of PSP.
Collapse
Affiliation(s)
- Kristen Whitney
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Won-Min Song
- Mount Sinai Center for Transformative Disease Modeling, Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Abhijeet Sharma
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Diana K Dangoor
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kurt Farrell
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Margaret M Krassner
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hadley W Ressler
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Thomas D Christie
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Shrishtee Kandoi
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ruth H Walker
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA
| | - Melissa J Nirenberg
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA
| | - Steven J Frucht
- The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Langone Health, New York, NY, USA
| | - Giulietta M Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Langone Health, New York, NY, USA
| | - Bin Zhang
- Mount Sinai Center for Transformative Disease Modeling, Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ana C Pereira
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA.
| | - John F Crary
- Department of Pathology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Neuropathology Brain Bank & Research Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA.
| |
Collapse
|
2
|
Li B, Zhao A, Tian T, Yang X. Mechanobiological insight into brain diseases based on mechanosensitive channels: Common mechanisms and clinical potential. CNS Neurosci Ther 2024; 30:e14809. [PMID: 38923822 PMCID: PMC11197048 DOI: 10.1111/cns.14809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND As physical signals, mechanical cues regulate the neural cells in the brain. The mechanosensitive channels (MSCs) perceive the mechanical cues and transduce them by permeating specific ions or molecules across the plasma membrane, and finally trigger a series of intracellular bioelectrical and biochemical signals. Emerging evidence supports that wide-distributed, high-expressed MSCs like Piezo1 play important roles in several neurophysiological processes and neurological disorders. AIMS To systematically conclude the functions of MSCs in the brain and provide a novel mechanobiological perspective for brain diseases. METHOD We summarized the mechanical cues and MSCs detected in the brain and the research progress on the functional roles of MSCs in physiological conditions. We then concluded the pathological activation and downstream pathways triggered by MSCs in two categories of brain diseases, neurodegenerative diseases and place-occupying damages. Finally, we outlined the methods for manipulating MSCs and discussed their medical potential with some crucial outstanding issues. RESULTS The MSCs present underlying common mechanisms in different brain diseases by acting as the "transportation hubs" to transduce the distinct signal patterns: the upstream mechanical cues and the downstream intracellular pathways. Manipulating the MSCs is feasible to alter the complicated downstream processes, providing them promising targets for clinical treatment. CONCLUSIONS Recent research on MSCs provides a novel insight into brain diseases. The common mechanisms mediated by MSCs inspire a wide range of therapeutic potentials targeted on MSCs in different brain diseases.
Collapse
Affiliation(s)
- Bolong Li
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
| | - An‐ran Zhao
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Tian Tian
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Xin Yang
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| |
Collapse
|
3
|
Whitney K, Song WM, Sharma A, Dangoor DK, Farrell K, Krassner MM, Ressler HW, Christie TD, Walker RH, Nirenberg MJ, Zhang B, Frucht SJ, Riboldi GM, Crary JF, Pereira AC. Single-cell transcriptomic and neuropathologic analysis reveals dysregulation of the integrated stress response in progressive supranuclear palsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567587. [PMID: 38014079 PMCID: PMC10680842 DOI: 10.1101/2023.11.17.567587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Progressive supranuclear palsy (PSP) is a sporadic neurodegenerative tauopathy variably affecting brainstem and cortical structures and characterized by tau inclusions in neurons and glia. The precise mechanism whereby these protein aggregates lead to cell death remains unclear. To investigate the contribution of these different cellular abnormalities to PSP pathogenesis, we performed single-nucleus RNA sequencing and analyzed 45,559 high quality nuclei targeting the subthalamic nucleus and adjacent structures from human post-mortem PSP brains with varying degrees of pathology compared to controls. Cell-type specific differential expression and pathway analysis identified both common and discrete changes in numerous pathways previously implicated in PSP and other neurodegenerative disorders. This included EIF2 signaling, an adaptive pathway activated in response to diverse stressors, which was the top activated pathway in vulnerable cell types. Using immunohistochemistry, we found that activated eIF2α was positively correlated with tau pathology burden in vulnerable brain regions. Multiplex immunofluorescence localized activated eIF2α positivity to hyperphosphorylated tau (p-tau) positive neurons and ALDH1L1-positive astrocytes, supporting the increased transcriptomic EIF2 activation observed in these vulnerable cell types. In conclusion, these data provide insights into cell-type-specific pathological changes in PSP and support the hypothesis that failure of adaptive stress pathways play a mechanistic role in the pathogenesis and progression of PSP.
Collapse
|
4
|
Procès A, Luciano M, Kalukula Y, Ris L, Gabriele S. Multiscale Mechanobiology in Brain Physiology and Diseases. Front Cell Dev Biol 2022; 10:823857. [PMID: 35419366 PMCID: PMC8996382 DOI: 10.3389/fcell.2022.823857] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/08/2022] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence suggests that mechanics play a critical role in regulating brain function at different scales. Downstream integration of mechanical inputs into biochemical signals and genomic pathways causes observable and measurable effects on brain cell fate and can also lead to important pathological consequences. Despite recent advances, the mechanical forces that influence neuronal processes remain largely unexplored, and how endogenous mechanical forces are detected and transduced by brain cells into biochemical and genetic programs have received less attention. In this review, we described the composition of brain tissues and their pronounced microstructural heterogeneity. We discuss the individual role of neuronal and glial cell mechanics in brain homeostasis and diseases. We highlight how changes in the composition and mechanical properties of the extracellular matrix can modulate brain cell functions and describe key mechanisms of the mechanosensing process. We then consider the contribution of mechanobiology in the emergence of brain diseases by providing a critical review on traumatic brain injury, neurodegenerative diseases, and neuroblastoma. We show that a better understanding of the mechanobiology of brain tissues will require to manipulate the physico-chemical parameters of the cell microenvironment, and to develop three-dimensional models that can recapitulate the complexity and spatial diversity of brain tissues in a reproducible and predictable manner. Collectively, these emerging insights shed new light on the importance of mechanobiology and its implication in brain and nerve diseases.
Collapse
Affiliation(s)
- Anthony Procès
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium.,Neurosciences Department, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Marine Luciano
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Yohalie Kalukula
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Laurence Ris
- Neurosciences Department, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Sylvain Gabriele
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| |
Collapse
|
5
|
Tracy TE, Madero-Pérez J, Swaney DL, Chang TS, Moritz M, Konrad C, Ward ME, Stevenson E, Hüttenhain R, Kauwe G, Mercedes M, Sweetland-Martin L, Chen X, Mok SA, Wong MY, Telpoukhovskaia M, Min SW, Wang C, Sohn PD, Martin J, Zhou Y, Luo W, Trojanowski JQ, Lee VMY, Gong S, Manfredi G, Coppola G, Krogan NJ, Geschwind DH, Gan L. Tau interactome maps synaptic and mitochondrial processes associated with neurodegeneration. Cell 2022; 185:712-728.e14. [PMID: 35063084 PMCID: PMC8857049 DOI: 10.1016/j.cell.2021.12.041] [Citation(s) in RCA: 161] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/20/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022]
Abstract
Tau (MAPT) drives neuronal dysfunction in Alzheimer disease (AD) and other tauopathies. To dissect the underlying mechanisms, we combined an engineered ascorbic acid peroxidase (APEX) approach with quantitative affinity purification mass spectrometry (AP-MS) followed by proximity ligation assay (PLA) to characterize Tau interactomes modified by neuronal activity and mutations that cause frontotemporal dementia (FTD) in human induced pluripotent stem cell (iPSC)-derived neurons. We established interactions of Tau with presynaptic vesicle proteins during activity-dependent Tau secretion and mapped the Tau-binding sites to the cytosolic domains of integral synaptic vesicle proteins. We showed that FTD mutations impair bioenergetics and markedly diminished Tau’s interaction with mitochondria proteins, which were downregulated in AD brains of multiple cohorts and correlated with disease severity. These multimodal and dynamic Tau interactomes with exquisite spatial resolution shed light on Tau’s role in neuronal function and disease and highlight potential therapeutic targets to block Tau-mediated pathogenesis. By combining APEX and AP-MS proteomic approaches, Tau interactome mapping reveals that Tau interactors are modified by neuronal activity and FTD mutations in human iPSC-derived neurons.
Collapse
Affiliation(s)
- Tara E Tracy
- Gladstone Institutes, San Francisco, CA 94158, USA; Buck Institute for Research on Aging, Novato, CA 94945, USA.
| | - Jesus Madero-Pérez
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Danielle L Swaney
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Timothy S Chang
- Department of Neurology, Movement Disorders Program and Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Michelle Moritz
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA
| | - Csaba Konrad
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Erica Stevenson
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ruth Hüttenhain
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Maria Mercedes
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lauren Sweetland-Martin
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xu Chen
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | | | - Sang-Won Min
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Chao Wang
- Gladstone Institutes, San Francisco, CA 94158, USA
| | | | | | - Yungui Zhou
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M Y Lee
- Center for Neurodegenerative Disease Research, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giovanni Coppola
- Department of Neurology, Movement Disorders Program and Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Daniel H Geschwind
- Department of Neurology, Movement Disorders Program and Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute of Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
6
|
Mechanosensing and the Hippo Pathway in Microglia: A Potential Link to Alzheimer's Disease Pathogenesis? Cells 2021; 10:cells10113144. [PMID: 34831369 PMCID: PMC8622675 DOI: 10.3390/cells10113144] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/01/2023] Open
Abstract
The activation of microglia, the inflammatory cells of the central nervous system (CNS), has been linked to the pathogenesis of Alzheimer’s disease and other neurodegenerative diseases. How microglia sense the changing brain environment, in order to respond appropriately, is still being elucidated. Microglia are able to sense and respond to the mechanical properties of their microenvironment, and the physical and molecular pathways underlying this mechanosensing/mechanotransduction in microglia have recently been investigated. The Hippo pathway functions through mechanosensing and subsequent protein kinase cascades, and is critical for neuronal development and many other cellular processes. In this review, we examine evidence for the potential involvement of Hippo pathway components specifically in microglia in the pathogenesis of Alzheimer’s disease. We suggest that the Hippo pathway is worth investigating as a mechanosensing pathway in microglia, and could be one potential therapeutic target pathway for preventing microglial-induced neurodegeneration in AD.
Collapse
|
7
|
Mohamed Asik R, Suganthy N, Aarifa MA, Kumar A, Szigeti K, Mathe D, Gulyás B, Archunan G, Padmanabhan P. Alzheimer's Disease: A Molecular View of β-Amyloid Induced Morbific Events. Biomedicines 2021; 9:biomedicines9091126. [PMID: 34572312 PMCID: PMC8468668 DOI: 10.3390/biomedicines9091126] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Amyloid-β (Aβ) is a dynamic peptide of Alzheimer’s disease (AD) which accelerates the disease progression. At the cell membrane and cell compartments, the amyloid precursor protein (APP) undergoes amyloidogenic cleavage by β- and γ-secretases and engenders the Aβ. In addition, externally produced Aβ gets inside the cells by receptors mediated internalization. An elevated amount of Aβ yields spontaneous aggregation which causes organelles impairment. Aβ stimulates the hyperphosphorylation of tau protein via acceleration by several kinases. Aβ travels to the mitochondria and interacts with its functional complexes, which impairs the mitochondrial function leading to the activation of apoptotic signaling cascade. Aβ disrupts the Ca2+ and protein homeostasis of the endoplasmic reticulum (ER) and Golgi complex (GC) that promotes the organelle stress and inhibits its stress recovery machinery such as unfolded protein response (UPR) and ER-associated degradation (ERAD). At lysosome, Aβ precedes autophagy dysfunction upon interacting with autophagy molecules. Interestingly, Aβ act as a transcription regulator as well as inhibits telomerase activity. Both Aβ and p-tau interaction with neuronal and glial receptors elevate the inflammatory molecules and persuade inflammation. Here, we have expounded the Aβ mediated events in the cells and its cosmopolitan role on neurodegeneration, and the current clinical status of anti-amyloid therapy.
Collapse
Affiliation(s)
- Rajmohamed Mohamed Asik
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
| | - Natarajan Suganthy
- Department of Nanoscience and Technology, Alagappa University, Karaikudi 630003, Tamil Nadu, India;
| | - Mohamed Asik Aarifa
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
| | - Arvind Kumar
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India;
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary; (K.S.); (D.M.)
- CROmed Translational Research Centers, 1094 Budapest, Hungary
| | - Domokos Mathe
- Department of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary; (K.S.); (D.M.)
- CROmed Translational Research Centers, 1094 Budapest, Hungary
- In Vivo Imaging Advanced Core Facility, Hungarian Center of Excellence for Molecular Medicine (HCEMM), 1094 Budapest, Hungary
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, 17176 Stockholm, Sweden
| | - Govindaraju Archunan
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
- Marudupandiyar College, Thanjavur 613403, Tamil Nadu, India
- Correspondence: (G.A.); (P.P.)
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Correspondence: (G.A.); (P.P.)
| |
Collapse
|
8
|
Castellani RJ. The Significance of Tau Aggregates in the Human Brain. Brain Sci 2020; 10:brainsci10120972. [PMID: 33322544 PMCID: PMC7763851 DOI: 10.3390/brainsci10120972] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022] Open
Abstract
Neurofibrillary degeneration has attracted the attention of neuroscientists as both a hallmark of the disease and a subject for experimentation for more than a century. Recent studies implicate phosphorylated tau (p-tau) directly in neurodegenerative disease pathogenesis, although the human data continue to raise questions. P-tau accumulates with age in a roughly hierarchical manner, but avoids abundance in the neocortex unless co-occurring with amyloid-β. Neurodegenerative tauopathies tend to have p-tau morphologies that differ from aging and Alzheimer’s disease. Tau isoforms (3R vs. 4R) have a tendency to vary with tauopathy phenotype for unknown reasons. Selective vulnerability to p-tau and spatial-temporal disconnect from amyloid-β are evident in aging. P-tau assessment at autopsy involves tissue decomposition, which may skew microanatomical observations toward limited biological meaning. Two major consensus guidelines for interpreting p-tau at autopsy emphasize the challenges of clinicopathologic correlation, and reinforce the observation that regional neurodegeneration is a better correlate of clinical signs than is proteinopathy. Despite the proliferation of interesting and novel theories related to tau-mediated pathogenesis, the weight of the human observations suggests that neurofibrillary degeneration is an epiphenomenal hallmark of aging and disease rather than an epicenter of neurotoxicity. This is consistent with numerous tau-targeted therapeutic strategies that have been unsuccessful to date.
Collapse
Affiliation(s)
- Rudy J. Castellani
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University, Morgantown, WV 26506, USA;
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
9
|
Hall CM, Moeendarbary E, Sheridan GK. Mechanobiology of the brain in ageing and Alzheimer's disease. Eur J Neurosci 2020; 53:3851-3878. [DOI: 10.1111/ejn.14766] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Chloe M. Hall
- Department of Mechanical Engineering University College London London UK
- School of Pharmacy and Biomolecular Sciences University of Brighton Brighton UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering University College London London UK
- Department of Biological Engineering Massachusetts Institute of Technology Cambridge MA USA
| | - Graham K. Sheridan
- School of Life Sciences Queens Medical Centre University of Nottingham Nottingham UK
| |
Collapse
|
10
|
Fischer I, Baas PW. Resurrecting the Mysteries of Big Tau. Trends Neurosci 2020; 43:493-504. [PMID: 32434664 DOI: 10.1016/j.tins.2020.04.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/28/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
Abstract
Tau, a microtubule-associated protein that modifies the dynamic properties and organization of microtubules in neurons and affects axonal transport, shows remarkable heterogeneity, with multiple isoforms (45-65 kDa) generated by alternative splicing. A high-molecular-weight (HMW) isoform (110 kDa) that contains an additional large exon termed 4a was discovered more than 25 years ago. This isoform, called Big tau, is expressed mainly in the adult peripheral nervous system (PNS), but also in adult neurons of the central nervous system (CNS) that extend processes into the periphery. Surprisingly little has been learned about Big tau since its initial characterization, leaving a significant gap in knowledge about how the dramatic switch to Big tau affects the properties of neurons in the context of development, disease, or injury. Here we review what was learned about the structure and distribution of Big tau in those earlier studies, and add contemporary insights to resurrect interest in the mysteries of Big tau and thereby set a path for future studies.
Collapse
Affiliation(s)
- Itzhak Fischer
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, 2900 Queen Lane, Philadelphia, PA 19129, USA.
| | - Peter W Baas
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, 2900 Queen Lane, Philadelphia, PA 19129, USA.
| |
Collapse
|
11
|
Huseby CJ, Bundschuh R, Kuret J. The role of annealing and fragmentation in human tau aggregation dynamics. J Biol Chem 2019; 294:4728-4737. [PMID: 30745358 DOI: 10.1074/jbc.ra118.006943] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/25/2019] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease pathogenesis is associated with the conversion of monomeric tau protein into filamentous aggregates. Because both toxicity and prion-like spread of pathogenic tau depend in part on aggregate size, the processes that underlie filament formation and size distribution are of special importance. Here, using a combination of biophysical and computational approaches, we investigated the fibrillation dynamics of the human tau isoform 2N4R. We found that tau filaments engage in a previously uncharacterized secondary process involving end-to-end annealing and that rationalization of empirical aggregation data composed of total protomer concentrations and fibril length distributions requires inclusion of this process along with filament fragmentation. We noted that annealing of 2N4R tau filaments is robust, with an intrinsic association rate constant of a magnitude similar to that mediating monomer addition and consistent with diffusion-mediated protein-protein interactions in the absence of long-range attractive forces. In contrast, secondary nucleation on the surface of tau filaments did not detectably contribute to tau aggregation dynamics. These results indicate that tau filament ends engage in a range of homotypic interactions involving monomers, oligomers, and filaments. They further indicate that, in the case of tau protein, fibril annealing and fragmentation along with primary nucleation and elongation are the major processes controlling filament size distribution.
Collapse
Affiliation(s)
| | - Ralf Bundschuh
- From the Interdisciplinary Biophysics Graduate Program.,Departments of Physics, Internal Medicine, and Chemistry and Biochemistry, and
| | - Jeff Kuret
- From the Interdisciplinary Biophysics Graduate Program, .,Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
12
|
Tang M, Harrison J, Deaton CA, Johnson GVW. Tau Clearance Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:57-68. [PMID: 32096028 DOI: 10.1007/978-981-32-9358-8_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Efficient quality control mechanisms are essential for a healthy, functional neuron. Recognition and degradation of misfolded, damaged, or potentially toxic proteins, is a crucial aspect of protein quality control. Tau is a protein that is highly expressed in neurons, and plays an important role in modulating a number of physiological processes. Maintaining appropriate levels of tau is key for neuronal health; hence perturbations in tau clearance mechanisms are likely significant contributors to neurodegenerative diseases such as Alzheimer's disease and frontotemporal lobar degeneration. In this chapter we will first briefly review the two primary degradative mechanisms that mediate tau clearance: the proteasome system and the autophagy-lysosome pathway. This will be followed by a discussion about what is known about the contribution of each of these pathways to tau clearance. We will also present recent findings on tau degradation through the endolysosomal system. Further, how deficits in these degradative systems may contribute to the accumulation of dysfunctional or toxic forms of tau in neurodegenerative conditions is considered.
Collapse
|
13
|
Breydo L, Redington JM, Uversky VN. Effects of Intrinsic and Extrinsic Factors on Aggregation of Physiologically Important Intrinsically Disordered Proteins. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 329:145-185. [PMID: 28109327 DOI: 10.1016/bs.ircmb.2016.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Misfolding and aggregation of proteins and peptides play an important role in a number of diseases as well as in many physiological processes. Many of the proteins that misfold and aggregate in vivo are intrinsically disordered. Protein aggregation is a complex multistep process, and aggregates can significantly differ in morphology, structure, stability, cytotoxicity, and self-propagation ability. The aggregation process is influenced by both intrinsic (e.g., mutations and expression levels) and extrinsic (e.g., polypeptide chain truncation, macromolecular crowding, posttranslational modifications, as well as interaction with metal ions, other small molecules, lipid membranes, and chaperons) factors. This review examines the effect of a variety of these factors on aggregation of physiologically important intrinsically disordered proteins.
Collapse
Affiliation(s)
- L Breydo
- Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| | - J M Redington
- Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - V N Uversky
- Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia.
| |
Collapse
|
14
|
Zhu S, Campbell JL, Chernushevich I, Le Blanc JCY, Wilson DJ. Differential Mobility Spectrometry-Hydrogen Deuterium Exchange (DMS-HDX) as a Probe of Protein Conformation in Solution. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:991-999. [PMID: 26965162 DOI: 10.1007/s13361-016-1364-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 06/05/2023]
Abstract
Differential mobility spectrometry (DMS) is an ion mobility technique that has been adopted chiefly as a pre-filter for small- to medium-sized analytes (<1 000 Da). With the exception of a handful of studies that employ an analogue of DMS-field asymmetric waveform ion mobility spectroscopy (FAIMS)-the application of DMS to intact biomacromolecules remains largely unexplored. In this work, we employ DMS combined with gas-phase hydrogen deuterium exchange (DMS-HDX) to probe the gas-phase conformations generated from proteins that were initially folded, partially-folded, and unfolded in solution. Our findings indicate that proteins with distinct structural features in solution exhibit unique deuterium uptake profiles as function of their optimal transmission through the DMS. Ultimately we propose that DMS-HDX can, if properly implemented, provide rapid measurements of liquid-phase protein structural stability that could be of use in biopharmaceuticals development. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Shaolong Zhu
- Chemistry Department, York University, Toronto, ON, M3J 1P3, Canada
| | | | | | | | - Derek J Wilson
- Chemistry Department, York University, Toronto, ON, M3J 1P3, Canada.
- Center for Research in Mass Spectrometry, Department of Chemistry, York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
15
|
Ghosh A, Giese KP. Calcium/calmodulin-dependent kinase II and Alzheimer's disease. Mol Brain 2015; 8:78. [PMID: 26603284 PMCID: PMC4657223 DOI: 10.1186/s13041-015-0166-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/13/2015] [Indexed: 11/10/2022] Open
Abstract
CaMKII is a remarkably complex protein kinase, known to have a fundamental role in synaptic plasticity and memory formation. Further, CaMKII has also been suggested to be a tau kinase. CaMKII dysregulation may therefore be a modulator of toxicity in Alzheimer's disease, a dementia characterised by aberrant calcium signalling, synapse and neuronal loss, and impaired memory. Here, we first examine the evidence for CaMKII dysregulation in Alzheimer's patients and draw parallels to findings in disease models which recapitulate key aspects of the disease. We then put forward the hypothesis that these changes critically contribute to neurodegeneration and memory impairment in Alzheimer's disease.
Collapse
Affiliation(s)
- Anshua Ghosh
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 5 Cutcombe Road, London, SE5 9RT, UK.
| | - Karl Peter Giese
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 5 Cutcombe Road, London, SE5 9RT, UK.
| |
Collapse
|
16
|
Zhu S, Shala A, Bezginov A, Sljoka A, Audette G, Wilson DJ. Hyperphosphorylation of intrinsically disordered tau protein induces an amyloidogenic shift in its conformational ensemble. PLoS One 2015; 10:e0120416. [PMID: 25767879 PMCID: PMC4359001 DOI: 10.1371/journal.pone.0120416] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/22/2015] [Indexed: 11/19/2022] Open
Abstract
Tau is an intrinsically disordered protein (IDP) whose primary physiological role is to stabilize microtubules in neuronal axons at all stages of development. In Alzheimer's and other tauopathies, tau forms intracellular insoluble amyloid aggregates known as neurofibrillary tangles, a process that appears in many cases to be preceded by hyperphosphorylation of tau monomers. Understanding the shift in conformational bias induced by hyperphosphorylation is key to elucidating the structural factors that drive tau pathology, however, as an IDP, tau is not amenable to conventional structural characterization. In this work, we employ a straightforward technique based on Time-Resolved ElectroSpray Ionization Mass Spectrometry (TRESI-MS) and Hydrogen/Deuterium Exchange (HDX) to provide a detailed picture of residual structure in tau, and the shifts in conformational bias induced by hyperphosphorylation. By comparing the native and hyperphosphorylated ensembles, we are able to define specific conformational biases that can easily be rationalized as enhancing amyloidogenic propensity. Representative structures for the native and hyperphosphorylated tau ensembles were generated by refinement of a broad sample of conformations generated by low-computational complexity modeling, based on agreement with the TRESI-HDX profiles.
Collapse
Affiliation(s)
- Shaolong Zhu
- Chemistry Department, York University, Toronto, ON, Canada
| | - Agnesa Shala
- Chemistry Department, York University, Toronto, ON, Canada
| | - Alexandr Bezginov
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Adnan Sljoka
- Department of Physics, Ryerson University, Toronto, ON, Canada
| | - Gerald Audette
- Chemistry Department, York University, Toronto, ON, Canada
| | - Derek J. Wilson
- Chemistry Department, York University, Toronto, ON, Canada
- Center for Research in Mass Spectrometry, Faculty of Science, York University, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
17
|
Kanaan NM, Himmelstein DS, Ward SM, Combs B, Binder LI. Tau Protein. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
18
|
Cook C, Stankowski JN, Carlomagno Y, Stetler C, Petrucelli L. Acetylation: a new key to unlock tau's role in neurodegeneration. ALZHEIMERS RESEARCH & THERAPY 2014; 6:29. [PMID: 25031639 PMCID: PMC4075151 DOI: 10.1186/alzrt259] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The identification of tau protein as a major constituent of neurofibrillary tangles spurred considerable effort devoted to identifying and validating pathways through which therapeutics may alleviate tau burden in Alzheimer's disease and related tauopathies, including chronic traumatic encephalopathy associated with sport- and military-related injuries. Most tau-based therapeutic strategies have previously focused on modulating tau phosphorylation, given that tau species present within neurofibrillary tangles are hyperphosphorylated on a number of different residues. However, the recent discovery that tau is modified by acetylation necessitates additional research to provide greater mechanistic insight into the spectrum of physiological consequences of tau acetylation, which may hold promise as a novel therapeutic target. In this review, we discuss recent findings evaluating tau acetylation in the context of previously accepted notions regarding tau biology and pathophysiology. We also examine the evidence demonstrating the neuroprotective and beneficial consequences of inhibiting histone deacetylase (HDAC)6, a tau deacetylase, including its effect on microtubule stabilization. We also discuss the rationale for pharmacologically modulating HDAC6 in tau-based pathologies as a novel therapeutic strategy.
Collapse
Affiliation(s)
- Casey Cook
- Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | - Yari Carlomagno
- Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | | |
Collapse
|
19
|
Himmelstein DS, Ward SM, Lancia JK, Patterson KR, Binder LI. Tau as a therapeutic target in neurodegenerative disease. Pharmacol Ther 2012; 136:8-22. [PMID: 22790092 DOI: 10.1016/j.pharmthera.2012.07.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 06/22/2012] [Indexed: 01/16/2023]
Abstract
Tau is a microtubule-associated protein thought to help modulate the stability of neuronal microtubules. In tauopathies, including Alzheimer's disease and several frontotemporal dementias, tau is abnormally modified and misfolded resulting in its disassociation from microtubules and the generation of pathological lesions characteristic for each disease. A recent surge in the population of people with neurodegenerative tauopathies has highlighted the immense need for disease-modifying therapies for these conditions, and new attention has focused on tau as a potential target for intervention. In the current work we summarize evidence linking tau to disease pathogenesis and review recent therapeutic approaches aimed at ameliorating tau dysfunction. The primary therapeutic tactics considered include kinase inhibitors and phosphatase activators, immunotherapies, small molecule inhibitors of protein aggregation, and microtubule-stabilizing agents. Although the evidence for tau-based treatments is encouraging, additional work is undoubtedly needed to optimize each treatment strategy for the successful development of safe and effective therapeutics.
Collapse
Affiliation(s)
- Diana S Himmelstein
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Tarry 8-754, 300 E. Superior St., Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
20
|
Abstract
Tauopathies are age-related neurodegenerative diseases that are characterized by the presence of aggregates of abnormally phosphorylated tau. As tau was originally discovered as a microtubule-associated protein, it has been hypothesized that neurodegeneration results from a loss of the ability of tau to associate with microtubules. However, tau has been found to have other functions aside from the promotion and stabilization of microtubule assembly. It is conceivable that such functions may be affected by the abnormal phosphorylation of tau and might have consequences for neuronal function or viability. This chapter provides an overview of tau structure, functions, and its involvement in neurodegenerative diseases.
Collapse
|
21
|
Breydo L, Uversky VN. Role of metal ions in aggregation of intrinsically disordered proteins in neurodegenerative diseases. Metallomics 2011; 3:1163-80. [PMID: 21869995 DOI: 10.1039/c1mt00106j] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neurodegenerative diseases constitute a set of pathological conditions originating from the slow, irreversible, and systematic cell loss within the various regions of the brain and/or the spinal cord. Depending on the affected region, the outcomes of the neurodegeneration are very broad and diverse, ranging from the problems with movements to dementia. Some neurodegenerative diseases are associated with protein misfolding and aggregation. Many proteins that misfold in human neurodegenerative diseases are intrinsically disordered; i.e., they lack a stable tertiary and/or secondary structure under physiological conditions in vitro. These intrinsically disordered proteins (IDPs) functionally complement ordered proteins, being typically involved in regulation and signaling. There is accumulating evidence that altered metal homeostasis may be related to the progression of neurodegenerative diseases. This review examines the effects of metal ion binding on the aggregation pathways of IDPs found in neurodegenerative diseases.
Collapse
Affiliation(s)
- Leonid Breydo
- Department of Molecular Medicine, College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd, MDC07, Tampa, Florida 33612, USA.
| | | |
Collapse
|
22
|
Chinnakkannu P, Samanna V, Cheng G, Ablonczy Z, Baicu CF, Bethard JR, Menick DR, Kuppuswamy D, Cooper G. Site-specific microtubule-associated protein 4 dephosphorylation causes microtubule network densification in pressure overload cardiac hypertrophy. J Biol Chem 2010; 285:21837-48. [PMID: 20436166 DOI: 10.1074/jbc.m110.120709] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In severe pressure overload-induced cardiac hypertrophy, a dense, stabilized microtubule network forms that interferes with cardiocyte contraction and microtubule-based transport. This is associated with persistent transcriptional up-regulation of cardiac alpha- and beta-tubulin and microtubule-stabilizing microtubule-associated protein 4 (MAP4). There is also extensive microtubule decoration by MAP4, suggesting greater MAP4 affinity for microtubules. Because the major determinant of this affinity is site-specific MAP4 dephosphorylation, we characterized this in hypertrophied myocardium and then assessed the functional significance of each dephosphorylation site found by mimicking it in normal cardiocytes. We first isolated MAP4 from normal and pressure overload-hypertrophied feline myocardium; volume-overloaded myocardium, which has an equal degree and duration of hypertrophy but normal functional and cytoskeletal properties, served as a control for any nonspecific growth-related effects. After cloning cDNA-encoding feline MAP4 and obtaining its deduced amino acid sequence, we characterized by mass spectrometry any site-specific MAP4 dephosphorylation. Solely in pressure overload-hypertrophied myocardium, we identified striking MAP4 dephosphorylation at Ser-472 in the MAP4 N-terminal projection domain and at Ser-924 and Ser-1056 in the assembly-promoting region of the C-terminal microtubule-binding domain. Site-directed mutagenesis of MAP4 cDNA was then used to switch each serine to non-phosphorylatable alanine. Wild-type and mutated cDNAs were used to construct adenoviruses; microtubule network density, stability, and MAP4 decoration were assessed in normal cardiocytes following an equivalent level of MAP4 expression. The Ser-924 --> Ala MAP4 mutant produced a microtubule phenotype indistinguishable from that seen in pressure overload hypertrophy, such that Ser-924 MAP4 dephosphorylation during pressure overload hypertrophy may be central to this cytoskeletal abnormality.
Collapse
Affiliation(s)
- Panneerselvam Chinnakkannu
- Cardiology Division, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina 29403, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Intrinsic Disorder in Proteins Associated with Neurodegenerative Diseases. PROTEIN FOLDING AND MISFOLDING: NEURODEGENERATIVE DISEASES 2008. [DOI: 10.1007/978-1-4020-9434-7_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
Uversky VN, Oldfield CJ, Dunker AK. Intrinsically disordered proteins in human diseases: introducing the D2 concept. Annu Rev Biophys 2008; 37:215-46. [PMID: 18573080 DOI: 10.1146/annurev.biophys.37.032807.125924] [Citation(s) in RCA: 1071] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intrinsically disordered proteins (IDPs) lack stable tertiary and/or secondary structures under physiological conditions in vitro. They are highly abundant in nature and their functional repertoire complements the functions of ordered proteins. IDPs are involved in regulation, signaling, and control, where binding to multiple partners and high-specificity/low-affinity interactions play a crucial role. Functions of IDPs are tuned via alternative splicing and posttranslational modifications. Intrinsic disorder is a unique structural feature that enables IDPs to participate in both one-to-many and many-to-one signaling. Numerous IDPs are associated with human diseases, including cancer, cardiovascular disease, amyloidoses, neurodegenerative diseases, and diabetes. Overall, intriguing interconnections among intrinsic disorder, cell signaling, and human diseases suggest that protein conformational diseases may result not only from protein misfolding, but also from misidentification, missignaling, and unnatural or nonnative folding. IDPs, such as alpha-synuclein, tau protein, p53, and BRCA1, are attractive targets for drugs modulating protein-protein interactions. From these and other examples, novel strategies for drug discovery based on IDPs have been developed. To summarize work in this area, we are introducing the D2 (disorder in disorders) concept.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Center for Computational Biology and Bioinformatics, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | |
Collapse
|
25
|
Abstract
Aggregation and subsequent development of protein deposition diseases originate from conformational changes in corresponding amyloidogenic proteins. The accumulated data support the model where protein fibrillogenesis proceeds via the formation of a relatively unfolded amyloidogenic conformation, which shares many structural properties with the pre-molten globule state, a partially folded intermediate first found during the equilibrium and kinetic (un)folding studies of several globular proteins and later described as one of the structural forms of natively unfolded proteins. The flexibility of this structural form is essential for the conformational rearrangements driving the formation of the core cross-beta structure of the amyloid fibril. Obviously, molecular mechanisms describing amyloidogenesis of ordered and natively unfolded proteins are different. For ordered protein to fibrillate, its unique and rigid structure has to be destabilized and partially unfolded. On the other hand, fibrillogenesis of a natively unfolded protein involves the formation of partially folded conformation; i.e., partial folding rather than unfolding. In this review recent findings are surveyed to illustrate some unique features of the natively unfolded proteins amyloidogenesis.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Biochemistry and Molecular Biology, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
26
|
Szőllősi E, Bokor M, Bodor A, Perczel A, Klement E, Medzihradszky KF, Tompa K, Tompa P. Intrinsic Structural Disorder of DF31, a Drosophila Protein of Chromatin Decondensation and Remodeling Activities. J Proteome Res 2008; 7:2291-9. [DOI: 10.1021/pr700720c] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Edit Szőllősi
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| | - Monika Bokor
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| | - Andrea Bodor
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| | - Andras Perczel
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| | - Eva Klement
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| | - Katalin F. Medzihradszky
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| | - Kalman Tompa
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| | - Peter Tompa
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary, Research Institute for Solid State Physics and Optics of Hungarian Academy of Sciences, Budapest, Hungary, Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary, Proteomics Research Group, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary, and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco,
| |
Collapse
|
27
|
Shahpasand K, Ahmadian S, Riazi GH. A possible mechanism for controlling processive transport by microtubule-associated proteins. Neurosci Res 2008; 61:347-50. [PMID: 18541318 DOI: 10.1016/j.neures.2008.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 04/16/2008] [Accepted: 04/25/2008] [Indexed: 01/07/2023]
Abstract
Molecular mechanisms of axonal transport have been evaluated by several investigators. It seems that microtubules (MTs) act as a track for the transport and microtubule-associated proteins (MAPs) seem to play as a regulating factor in it. In order to transport MTs must move in the radial direction to make room for a vesicle and when the cargo passes, return to the previous position for the maintenance of neuronal structure. An inhibitor factor against the radial movement is the steric constraints resulted from presence of MAPs. In fact, inter-microtubular spaces (IMS) in the neuronal processes are resulted from the space-making role of the MAPs. Since the IMS must be locally altered to make enough room for a vesicle, it seems relevant to imagine some mechanisms that control the steric constraints for an efficient vesicular transport. Here we juxtapose the older findings and the recent ones to investigate the possible effects of MAPs on the processive transport.
Collapse
Affiliation(s)
- Kourosh Shahpasand
- Institute of Biochemistry & Biophysics, University of Tehran, Tehran, Iran.
| | | | | |
Collapse
|
28
|
Froelich-Fabre S, Skoglund L, Ostojic J, Kilander L, Lindau M, Glaser A, Basun H, Lannfelt L. Clinical and molecular aspects of frontotemporal dementia. NEURODEGENER DIS 2006; 1:218-24. [PMID: 16908993 DOI: 10.1159/000080989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease and next to Alzheimer's disease and vascular dementia, the third most common cause of early-onset progressive dementia. FTD leads to neurodegeneration in the frontal and temporal neocortex and usually encompasses both sides of the frontal and anterior temporal lobes. Psychologically, FTD is characterized by personality changes such as lack of insight, inappropriate behaviour, disinhibition, apathy, executive disabilities and a decline in cognitive functions, with large clinical and neuropathological variations among cases. Neuropathological characteristics include gliosis or microvacuolation of cortical nerve cells. Inclusions staining for tau protein and/or ubiquitin are also common findings. Both sporadic and hereditary forms of FTD have been identified and 30-50% of the FTD cases have a familial background. So far, at least three genetic loci for FTD have been identified, at human chromosomes 3, 9 and 17 in familial forms of the disease. A large number of the familial forms have been linked to chromosome 17q21 and referred to as frontotemporal dementia and Parkinsonism linked to chromosome 17. In the majority of these families, pathogenic mutations in the tau gene were identified. However, tau mutations seem to be a rare cause of disease in the general FTD population. Thus, other genes and/or environmental factors are yet to be identified, which will give further clues to this complex and heterogeneous disorder.
Collapse
Affiliation(s)
- Susanne Froelich-Fabre
- Department of Public Health and Caring Science, Division of Geriatrics, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Rankin CA, Sun Q, Gamblin TC. Pseudo-phosphorylation of tau at Ser202 and Thr205 affects tau filament formation. ACTA ACUST UNITED AC 2005; 138:84-93. [PMID: 15913837 DOI: 10.1016/j.molbrainres.2005.04.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 01/19/2005] [Accepted: 04/13/2005] [Indexed: 01/25/2023]
Abstract
The microtubule-associated protein tau aggregates into insoluble filaments in numerous neurodegenerative diseases, most common of which is Alzheimer's disease. Tau aggregation in Alzheimer's disease appears to follow a continuum from soluble monomer to an end point of insoluble extracellular tangles with a strong correlation between the amount of fibrillar tau and dementia. The phosphorylation of amino acids S202 and T205 in the tau molecule is recognized by the phosphorylation-specific monoclonal antibody, AT8, and has been observed by a number of researchers to be an early step in the progression of monomer to filaments. In addition, these amino acids are located in a proline-rich region containing a set of five phosphorylation sites (one being S202), that when phosphorylated, were reported to alter several properties of tau, including filament formation. Considering these observations, we have investigated the role of S202 and T205 phosphorylation in the in vitro polymerization of tau. Pseudo-phosphorylation mutants were constructed by site-directed mutagenesis in which amino acids S202 and T205 were changed to negatively charged glutamic acids mimicking post-translational phosphorylation. These pseudo-phosphorylated, mutant tau proteins were then assayed in vitro for changes in structure, polymerization into filaments, and microtubule binding. Phosphorylation at the AT8 site does not appear to influence either SDS-resistant structure nor microtubule binding. However, in regard to filament formation, phosphorylation at S202 appears to enhance polymerization; and phosphorylation at both sites not only enhances polymerization but also makes filament formation more sensitive to small changes in tau concentration.
Collapse
Affiliation(s)
- Carolyn A Rankin
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA.
| | | | | |
Collapse
|
30
|
Tamada Y, Nakajima E, Nakajima T, Shearer TR, Azuma M. Proteolysis of neuronal cytoskeletal proteins by calpain contributes to rat retinal cell death induced by hypoxia. Brain Res 2005; 1050:148-55. [PMID: 15979593 DOI: 10.1016/j.brainres.2005.05.048] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2005] [Revised: 05/17/2005] [Accepted: 05/18/2005] [Indexed: 11/29/2022]
Abstract
Our previous studies in retina on the mechanism for hypoxia-induced cell death suggested activation of a class of calcium-activated proteases known as calpains. This conclusion was based on data showing proteolysis of a calpain substrate alpha-spectrin, autolysis of activated calpain, and reduction of cell damage by calpain inhibitor SJA6017. Less is known about changes in downstream pathways after calpain activation. Thus, the purpose of the present investigation was to measure proteolysis of neuronal cytoskeletal proteins and apoptotic cell signaling factors during hypoxia-induced retinal cell death. Rat retinas were incubated in RPMI medium with glucose and 95% O2/5% CO2 to supply sufficient oxygen for retinal cell survival. Hypoxia was induced with 95% N2/5% CO2 without glucose. Immunoblotting was used to detect activation of calpain and proteolysis of substrates. Amounts of mRNA for calpain 1 and 2 were determined by quantitative PCR. Twelve times more calpain 2 mRNA than calpain 1 was present in retinas. Activation of calpain 2 and production of a calpain-specific alpha-spectrin breakdown product at 150 kDa were confirmed in hypoxic retinas. Further, pro-caspase-3 at 32 kDa was proteolyzed to a fragment at 30 kDa, tau protein was lost, and p35 was proteolyzed to p25 suggesting prolonged activation of cdk5. SJA6017 partially inhibited the production of these fragments. During hypoxia in rat retinas, calpains may be major proteases causing breakdown of neuronal proteins involved in apoptotic cell death. Calpain inhibitor SJA6017 may have potential for testing as a therapeutic agent against retinal pathologies such those caused by glaucoma, although future studies such as testing in in vivo animal models are required.
Collapse
Affiliation(s)
- Yoshiyuki Tamada
- Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Corporation Limited, Beaverton, OR 97006, USA
| | | | | | | | | |
Collapse
|
31
|
Ksiezak-Reding H, Wall JS. Characterization of paired helical filaments by scanning transmission electron microscopy. Microsc Res Tech 2005; 67:126-40. [PMID: 16104001 DOI: 10.1002/jemt.20188] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Paired helical filaments (PHFs) are abnormal twisted filaments composed of hyperphosphorylated tau protein. They are found in Alzheimer's disease and other neurodegenerative disorders designated as tauopathies. They are a major component of intracellular inclusions known as neurofibrillary tangles (NFTs). The objective of this review is to summarize various structural studies of PHFs in which using scanning transmission electron microscopy (STEM) has been particularly informative. STEM provides shape and mass per unit length measurements important for studying ultrastructural aspects of filaments. These include quantitative comparisons between dispersed and aggregated populations of PHFs as well as comparative studies of PHFs in Alzheimer's disease and other neurodegenerative disorders. Other approaches are also discussed if relevant or complementary to studies using STEM, e.g., application of a novel staining reagent, Nanovan. Our understanding of the PHF structure and the development of PHFs into NFTs is presented from a historical perspective. Others goals are to describe the biochemical and ultrastructural complexity of authentic PHFs, to assess similarities between authentic and synthetic PHFs, and to discuss recent advances in PHF modeling.
Collapse
Affiliation(s)
- Hanna Ksiezak-Reding
- Neuroinflammation Research Laboratories, Department of Psychiatry, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | | |
Collapse
|
32
|
Hall GF, Yao J. Modeling tauopathy: a range of complementary approaches. Biochim Biophys Acta Mol Basis Dis 2004; 1739:224-39. [PMID: 15615641 DOI: 10.1016/j.bbadis.2004.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2004] [Accepted: 10/05/2004] [Indexed: 11/24/2022]
Abstract
The large group of neurodegenerative diseases which feature abnormal metabolism and accumulation of tau protein (tauopathies) characteristically produce a multiplicity of cellular and systemic abnormalities in human patients. Understanding the complex pathogenetic mechanisms by which abnormalities in tau lead to systemic neurofibrillary degenerative disease requires the construction and use of model experimental systems in which the behavior of human tau can be analyzed under controlled conditions. In this paper, we survey the ways in which in vitro, cellular and whole-animal models of human tauopathy are being used to add to our knowledge of the pathogenetic mechanisms underlying these conditions. In particular, we focus on the complementary advantages and limitations of various approaches to constructing tauopathy models presently in use with respect to those of murine transgenic tauopathy models.
Collapse
Affiliation(s)
- Garth F Hall
- Department of Biological Sciences, University of Massachusetts, Lowell, Massachusetts 01854, USA.
| | | |
Collapse
|
33
|
Mukhopadhyay R, Kumar S, Hoh JH. Molecular mechanisms for organizing the neuronal cytoskeleton. Bioessays 2004; 26:1017-25. [PMID: 15351972 DOI: 10.1002/bies.20088] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Neurofilaments and microtubules are important components of the neuronal cytoskeleton. In axons or dendrites, these filaments are aligned in parallel arrays, and separated from one another by nonrandom distances. This distinctive organization has been attributed to cross bridges formed by NF side arms or microtubule-associated proteins. We recently proposed a polymer-brush-based mechanism for regulating interactions between neurofilaments and between microtubules. In this model, the side arms of neurofilaments and the projection domains of microtubule-associated proteins are highly unstructured and exert long-range repulsive forces that are largely entropic in origin; these forces then act to organize the cytoskeleton in axons and dendrites. Here, we review the biochemical, biophysical, genetic and cell biological data for the polymer-brush and cross-bridging models. We explore how the data traditionally used to support cross bridging may be reconciled with a polymer-brush mechanism and compare the implications of recent experimental insights into axonal transport and physiology for each model.
Collapse
|
34
|
Uversky VN, Fink AL. Conformational constraints for amyloid fibrillation: the importance of being unfolded. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2004; 1698:131-53. [PMID: 15134647 DOI: 10.1016/j.bbapap.2003.12.008] [Citation(s) in RCA: 796] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2003] [Revised: 12/01/2003] [Accepted: 12/01/2003] [Indexed: 02/07/2023]
Abstract
Recent reports give strong support to the idea that amyloid fibril formation and the subsequent development of protein deposition diseases originate from conformational changes in corresponding amyloidogenic proteins. In this review, recent findings are surveyed to illustrate that protein fibrillogenesis requires a partially folded conformation. This amyloidogenic conformation is relatively unfolded, and shares many structural properties with the pre-molten globule state, a partially folded intermediate frequently observed in the early stages of protein folding and under some equilibrium conditions. The inherent flexibility of such an intermediate is essential in allowing the conformational rearrangements necessary to form the core cross-beta structure of the amyloid fibril.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA.
| | | |
Collapse
|
35
|
|
36
|
Simić G, Diana A, Hof PR. Phosphorylation pattern of tau associated with distinct changes of the growth cone cytoskeleton. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2003; 32:33-48. [PMID: 12827970 DOI: 10.1007/978-3-642-55557-2_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- G Simić
- Department of Neuroscience, Croatian Institute for Brain Research, Zagreb University Medical School, Salata 12, Zagreb 10000, Croatia
| | | | | |
Collapse
|
37
|
Higuchi M, Lee VMY, Trojanowski JQ. Tau and axonopathy in neurodegenerative disorders. Neuromolecular Med 2003; 2:131-50. [PMID: 12428808 DOI: 10.1385/nmm:2:2:131] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2002] [Accepted: 06/10/2002] [Indexed: 01/25/2023]
Abstract
The microtubule (MT)-associated protein (MAP) tau in neurons has been implicated as a significant factor in the axonal growth, development of neuronal polarity, and the maintenance of MT dynamics. Tau is localized to the axon, and is known to promote MT assembly and to stabilize axonal MTs. These functions of tau are primarily regulated by the activities of protein kinases and phosphatases. In Alzheimer's disease and other neurodegenerative disorders, abundant filamentous tau inclusions are found to be major neuropathological characteristics of these diseases. Both somato-dendritic and axonal tau lesions appear to be closely associated with axonal disruption. Furthermore, recent discoveries of pathogenic mutations on the tau gene suggest that abnormalities of tau alone are causative of neurodegeneration. Finally, analyses of transgenic mice that express human tau proteins have enabled in vivo quantitative assessments of axonal functions and have provided information about mechanistic relationships between pathological alteration of tau and axonal degeneration.
Collapse
|
38
|
Yamamoto H, Yamauchi E, Taniguchi H, Ono T, Miyamoto E. Phosphorylation of microtubule-associated protein tau by Ca2+/calmodulin-dependent protein kinase II in its tubulin binding sites. Arch Biochem Biophys 2002; 408:255-62. [PMID: 12464279 DOI: 10.1016/s0003-9861(02)00556-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The paired helical filaments (PHF) found in Alzheimer's disease (AD) brain are composed mainly of the hyperphosphorylated form of microtubule-associated protein tau (PHF-tau). It is well known that tau is a good in vitro substrate for Ca(2+)/calmodulin-dependent protein kinase II (CaM kinase II). To establish the phosphorylation sites, the longest human tau (hTau40) was bacterially expressed and phosphorylated by CaM kinase II, followed by digestion with lysyl endoprotease. The digests were subjected to liquid chromatography/mass spectrometry. We found that 5 of 22 identified peptides were phosphorylated. From the tandem mass spectrometry, two phosphorylation sites (serines 262 and 356) were identified in the tubulin binding sites. When tau was phosphorylated by CaM kinase II, the binding of tau to taxol-stabilized microtubules was remarkably impaired. As both serines 262 and 356 are reportedly phosphorylated in PHF-tau, CaM kinase II may be involved in hyperphosphorylation of tau in AD brain.
Collapse
Affiliation(s)
- Hideyuki Yamamoto
- Department of Pharmacology, Kumamoto University School of Medicine, Kumamoto, Japan.
| | | | | | | | | |
Collapse
|
39
|
Gold M. Tau therapeutics for Alzheimer's disease: the promise and the challenges. J Mol Neurosci 2002; 19:331-4. [PMID: 12540060 DOI: 10.1385/jmn:19:3:329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The pathological diagnosis of Alzheimer's disease (AD) depends on the presence of plaques consisting of the beta-amyloid peptide as well as neurofibrillary tangles consisting of paired helical filaments (PHFs) of the tau (tau) protein. The role of each type of pathology in the pathogenesis and progression of AD remains unclear. Previous hypotheses suggested that these two processes were independent, whereas more recent data suggest that there may be a bidirectional interaction between these two pathological processes. The identification of the neurotoxic effects of beta-amyloid and the discovery of mutations responsible for early-onset Alzheimer's disease (EOAD) and their linkage to beta-amyloid overproduction, has made the amyloid hypothesis of AD the predominant influence for therapeutic targets. Several approaches have emerged from preclinical testing and have entered early phases of clinical developments. The recent identification of tau mutations and their linkage to progressive neurodegenerative disorders provides a counterbalancing influence on the search for therapeutic targets for AD. Therapeutic approaches that are targeted to either beta-amyloid or tau share certain features at the level of pharmacology and will face many of the same challenges as they progress through drug development paradigms. The aim of this article is to provide a brief overview of some of the commonalities and the challenges faced by tau-related therapeutic strategies. The issues discussed in this article are not exhaustively dealt with in either scope or detail.
Collapse
Affiliation(s)
- Michael Gold
- Global Clinical Research and Development, CNS/Analgesia, Johnson & Johnson Pharmaceutical Research and Development, Titusville, NJ 08560, USA.
| |
Collapse
|
40
|
Goux WJ. The conformations of filamentous and soluble tau associated with Alzheimer paired helical filaments. Biochemistry 2002; 41:13798-806. [PMID: 12427043 DOI: 10.1021/bi016079h] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Paired helical filaments (PHF) occur in Alzheimer's diseased brains and are known to be composed of the microtubule-associated protein, tau. In the present report, circular dichroism (CD) spectroscopy and transmission electron microscopy (TEM) were used to characterize PHF suspended in Tris-buffered saline (TBS), sodium acetate buffer, and water. In TBS the CD spectrum of PHF was observed to have a spectral pattern consistent with 31-37% alpha-helix, 15-20% beta-sheet, 20-23% turn, and 26-29% unordered structure. The TBS sample was found to undergo a cooperative thermal transition between 70 and 75 degrees C, consistent with the changes observed in filament morphology, and it suggests that filamentous tau in the PHF (PHF-tau) makes a substantial contribution to the overall CD. Observed changes in the CD spectrum following removal of PHF by centrifugation suggest that PHF-tau possesses a higher fraction of alpha-helical structure than soluble tau. In acetate buffer, where only straight filaments were observed, the CD was consistent with a marked decrease in the fraction of alpha-helix and an increase in the fraction of beta-sheet relative to the sample in TBS. In water, where only rudimentary filaments remain, the CD was consistent with a Type II or II' beta-turn conformation. Only noncooperative thermal transitions were observed for the PHF samples in acetate buffer and water, consistent with the presence of a heterogeneous population of folded structures. Taken cumulatively, the results are consistent with immunological data showing the presence of folded forms of tau and suggest that phosphorylation or nonproteinaceous components are able to induce conformations of tau other than the random coil conformation previously reported for cloned or purified human tau.
Collapse
Affiliation(s)
- Warren J Goux
- Department of Chemistry, The University of Texas at Dallas, P.O. Box 830688, Richardson, Texas 75080, USA.
| |
Collapse
|
41
|
Cassimeris L, Spittle C. Regulation of microtubule-associated proteins. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 210:163-226. [PMID: 11580206 DOI: 10.1016/s0074-7696(01)10006-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microtubule-associated proteins (MAPs) function to regulate the assembly dynamics and organization of microtubule polymers. Upstream regulation of MAP activities is the major mechanism used by cells to modify and control microtubule assembly and organization. This review summarizes the functional activities of MAPs found in animal cells and discusses how these MAPs are regulated. Mechanisms controlling gene expression, isoform-specific expression, protein localization, phosphorylation, and degradation are discussed. Additional regulatory mechanisms include synergy or competition between MAPs and the activities of cofactors or binding partners. For each MAP it is likely that regulation in vivo reflects a composite of multiple regulatory mechanisms.
Collapse
Affiliation(s)
- L Cassimeris
- Department of Biological Sciences, Lehigh University Bethlehem, Pennsylvania 18015, USA
| | | |
Collapse
|
42
|
Bélanger D, Farah CA, Nguyen MD, Lauzon M, Cornibert S, Leclerc N. The projection domain of MAP2b regulates microtubule protrusion and process formation in Sf9 cells. J Cell Sci 2002; 115:1523-39. [PMID: 11896199 DOI: 10.1242/jcs.115.7.1523] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The expression of microtubule-associated protein 2 (MAP2), developmentally regulated by alternative splicing, coincides with neurite outgrowth. MAP2 proteins contain a microtubule-binding domain (C-terminal) that promotes microtubule assembly and a poorly characterized domain, the projection domain(N-terminal), extending at the surface of microtubules. MAP2b differs from MAP2c by an additional sequence of 1372 amino acids in the projection domain. In this study, we examined the role of the projection domain in the protrusion of microtubules from the cell surface and the subsequent process formation in Sf9 cells. In this system, MAP2b has a lower capacity to induce process formation than MAP2c. To investigate the role of the projection domain in this event, we expressed truncated forms of MAP2b and MAP2c that have partial or complete deletion of their projection domain in Sf9 cells. Our results indicate that process formation is induced by the microtubule-binding domain of these MAP2 proteins and is regulated by their projection domain. Furthermore, the microtubule-binding activity of MAP2b and MAP2c truncated forms as well as the structural properties of the microtubule bundles induced by them do not seem to be the only determinants that control the protrusion of microtubules from the cell surface in Sf9 cells. Rather, our data suggest that microtubule protrusion and process formation are regulated by intramolecular interactions between the projection domain and its microtubule-binding domain in MAP2b.
Collapse
Affiliation(s)
- Dave Bélanger
- Département de pathologie et biologie cellulaire, Université de Montréal, Montréal, Québec, Canada, H3T 1J8
| | | | | | | | | | | |
Collapse
|
43
|
Abraha A, Ghoshal N, Gamblin TC, Cryns V, Berry RW, Kuret J, Binder LI. C-terminal inhibition of tau assembly in vitro and in Alzheimer's disease. J Cell Sci 2000; 113 Pt 21:3737-45. [PMID: 11034902 DOI: 10.1242/jcs.113.21.3737] [Citation(s) in RCA: 215] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease (AD) is, in part, defined by the polymerization of tau into paired helical and straight filaments (PHF/SFs) which together comprise the fibrillar pathology in degenerating brain regions. Much of the tau in these filaments is modified by phosphorylation. Additionally, a subset also appears to be proteolytically truncated, resulting in the removal of its C terminus. Antibodies that recognize tau phosphorylated at S(396/404)or truncated at E(391) do not stain control brains but do stain brain sections very early in the disease process. We modeled these phosphorylation and truncation events by creating pseudo-phosphorylation and deletion mutants derived from a full-length recombinant human tau protein isoform (ht40) that contains N-terminal exons 2 and 3 and all four microtubule-binding repeats. In vitro assembly experiments demonstrate that both modifications greatly enhance the rates of tau filament formation and that truncation increases the mass of polymer formed, as well. Removal of as few as 12 or as many as 121 amino acids from the C terminus of tau greatly increases the rate and extent of tau polymerization. However, deletion of an additional 7 amino acids, (314)DLSKVTS(320), from the third microtubule-binding repeat results in the loss of tau's ability to form filaments in vitro. These results suggest that only part of the microtubule-binding domain (repeats 1, 2 and a small portion of 3) is crucial for tau polymerization. Moreover, the C terminus of tau clearly inhibits the assembly process; this inhibition can be partially reversed by site-specific phosphorylation and completely removed by truncation events at various sites from S(320) to the end of the molecule.
Collapse
Affiliation(s)
- A Abraha
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, IL 60611, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Eidenmüller J, Fath T, Hellwig A, Reed J, Sontag E, Brandt R. Structural and functional implications of tau hyperphosphorylation: information from phosphorylation-mimicking mutated tau proteins. Biochemistry 2000; 39:13166-75. [PMID: 11052669 DOI: 10.1021/bi001290z] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Abnormal tau-immunoreactive filaments are a hallmark of tauopathies, including Alzheimer's disease (AD). A higher phosphorylation ("hyperphosphorylation") state of tau protein may represent a critical event. To determine the potential role of tau hyperphosphorylation in these disorders, mutated tau proteins were produced where serine/threonine residues known to be highly phosphorylated in tau filaments isolated from AD patients were substituted for glutamate to simulate a paired helical filament (PHF)-like tau hyperphosphorylation. We demonstrate that, like hyperphosphorylation, glutamate substitutions induce compact structure elements and SDS-resistant conformational domains in tau protein. Hyperphosphorylation-mimicking glutamate-mutated tau proteins display a complete functional loss in its ability to promote microtubule nucleation which can partially be overcome by addition of the osmolyte trimethylamine N-oxide (TMAO), which is similar to phosphorylated tau. In addition, glutamate-mutated tau proteins fail to interact with the dominant brain protein phosphatase 2A isoform ABalphaC, and exhibit a reduced ability to assemble into filaments. Interestingly, wild-type tau and phosphorylation-mimicking tau similarly bind to microtubules when added alone, but the mutated tau is almost completely displaced from the microtubule surface by equimolar concentrations of wild-type tau. The data indicate that glutamate-mutated tau proteins provide a useful model for analyzing the functional consequences of tau hyperphosphorylation. They suggest that several mechanisms contribute to the abnormal tau accumulation observed during tauopathies, in particular a selective displacement of hyperphosphorylated tau from microtubules, a functional loss in promoting microtubule nucleation, and a failure to interact with phosphatases.
Collapse
Affiliation(s)
- J Eidenmüller
- Department of Neurobiology, IZN, University of Heidelberg, INF 345, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
1. The potential functions of the microtubule-associated protein tau have been expanded by the recent demonstration of its interaction with the plasma membrane. Since the association of tau with microtubules is regulated by phosphorylation, herein we examine whether or not the association of tau with the plasma membrane is also regulated by phosphorylation. 2. A range of tau isoforms migrating from 46 to 64 kDa was associated with crude particulate fractions derived from SH-SY-5Y human neuroblastoma cells, and were retained during the initial stages of plasma membrane purification. During the extensive washing utilized in purification of the plasma membrane, portions of each of these isoforms were depleted from the resultant purified membrane. Immunoblot analysis with phospho-dependent and -independent antibodies revealed selective depletion of phospho isoforms during membrane washing. This effect was more pronounced for the slowest-migrating (64-kDa) tau isoform. 3. This putative influence of phosphorylation on the association of tau with the plasma membrane was further probed by transfection of SH-SY-5Y human neuroblastoma cells with a tau construct that could associate with the plasma membrane but not with microtubules. Treatment with phorbol ester or calcium ionophore, both of which increased phospho-tau levels within the cytosol and plasma membrane, was accompanied by the dissociation of this tau construct from the membrane. 4. These data indicate that phosphorylation regulates the association with the plasma membrane. Dissociation from the membrane by phosphorylation may place tau at risk for hyperphosphorylation and ultimate PHF formation in a manner previously considered for tau dissociated from microtubules.
Collapse
Affiliation(s)
- F J Ekinci
- Center for Cellular Neurobiology and Neurodegeneration Research, Department of Biological Sciences, University of Massachusetts at Lowell, 01854, USA
| | | |
Collapse
|
46
|
Janke C, Beck M, Holzer M, Bigl V, Arendt T. Analysis of the molecular heterogeneity of the microtubule-associated protein tau by two-dimensional electrophoresis and RT-PCR. BRAIN RESEARCH. BRAIN RESEARCH PROTOCOLS 2000; 5:231-42. [PMID: 10906488 DOI: 10.1016/s1385-299x(00)00018-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The microtubule-associated protein tau is a member of a group of proteins, promoting assembly and stabilization of microtubules. In several tauopathic neurodegenerative disorders, namely Alzheimer's and Pick's disease and frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP 17) this protein is converted into fibrilar polymers which form the component of insoluble proteanous deposits such as neurofibrillary tangles. The formation of these fibrils is believed to interrupt the physiological function of neurons resulting in degeneration and cell death. Tau protein exists as a family of heterogeneous isoforms derived by both, differential splicing of tau-mRNA and posttranslational modification of the protein. Since the role of the different isoforms during the process of neurodegeneration is not well understood and as their balance might be altered in some cases of tauopathies (Spillantini et al., Proc. Natl. Acad. Sci. USA 1998;95:7737-7741), the detailed analysis of the molecular heterogeneity gained outstanding interest. The method presented here allows the analysis of both, differential splicing and phosphorylation of tau protein by the application of two-dimensional (2D) electrophoresis and Western blot detection. Tau protein isoforms could be identified from the 2D pattern of dephosphorylated tau in concordance with the results of tau-mRNA analysis by RT-PCR. The protocol presented was successfully applied to analysis of tau isoforms of human brain (Janke et al., FEBS Lett. 1996;379:222-226) and of several species, revealing a phylogenetic correlation of tau protein patterns in mammals (Janke et al., Mol. Brain Res. 1999;68:119-128). The present paper provides a detailed description of the technique and discusses its prospects and limits.
Collapse
Affiliation(s)
- C Janke
- Department of Neuroanatomy, University of Leipzig, Paul Flechsig Institute of Brain Research, Jahnallee 59, 04109, Leipzig, Germany
| | | | | | | | | |
Collapse
|
47
|
Torreilles F, Roquet F, Granier C, Pau B, Mourton-Gilles C. Binding specificity of monoclonal antibody AD2: influence of the phosphorylation state of tau. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 78:181-5. [PMID: 10891598 DOI: 10.1016/s0169-328x(00)00073-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Using recombinant human tau protein phosphorylated by a brain extract and the glycogen synthase kinase-3beta in the absence or the presence of heparin, we showed that phosphorylation-dependent antibody AD2 recognition only requires phosphorylated Ser-396. By the Spot multiple peptide synthesis method, we showed that Tyr-394, Ser(P)-396 and Pro-397 are critical for AD2 binding. A decrease in the binding of AD2 was observed with increasing phosphorylation of residues in the vicinity of Ser(P)-396.
Collapse
Affiliation(s)
- F Torreilles
- CNRS UMR 5094 Institut de Biotechnologie en Immunoanalyse et Pharmacologie UFR Pharmacie, 15 Av. Charles Flahault, 34060 Montpellier Cedex 2, France
| | | | | | | | | |
Collapse
|
48
|
Gordon-Krajcer W, Yang L, Ksiezak-Reding H. Conformation of paired helical filaments blocks dephosphorylation of epitopes shared with fetal tau except Ser199/202 and Ser202/Thr205. Brain Res 2000; 856:163-75. [PMID: 10677623 DOI: 10.1016/s0006-8993(99)02391-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
To determine if the high phosphate content of paired helical filaments (PHFs) in Alzheimer's disease (AD) is a result of limited access to filament phosphorylation sites, we studied in vitro dephosphorylation of intact PHFs, PHFs with filamentous structure abolished by formic acid treatment (PHF(FA)) and fetal human tau protein. Samples were treated with alkaline phosphatase for up to 24 h at 37 degrees C and then immunoblotted with eight well characterized tau antibodies, that recognize two phosphorylation-insensitive sites and six phosphorylation-sensitive epitopes at Thr181, Ser199/202, Ser202/Thr205, Thr231, Ser262/356 and Ser396/404. Intact PHFs were effectively dephosphorylated only at the two N-terminal epitopes Ser199/202 and Ser202/Thr205, with little change in electrophoretic mobility. In contrast, PHF(FA) were dephosphorylated at all epitopes, with particular effectiveness at those in the C-terminus and with significant increase in electrophoretic mobility. The fetal tau epitopes were effectively dephosphorylated except at Thr181 and Thr231 with marked increase in mobility. The extent of dephosphorylation of PHF(FA) was equal or more effective than in fetal tau, except for Thr181 that was minimally dephosphorylated in both proteins. The results indicate that intact PHFs, but not PHF(FA) or fetal tau display differential dephosphorylation of the N- and C-terminal epitopes. The results confirm that the filamentous conformation may significantly contribute to hyperphosphorylation of PHFs in the C-terminus. The filamentous conformation, however, does not limit access to two N-terminal epitopes Ser199/202 and Ser202/Thr205. The access to these sites in AD may be limited by other factors, e.g., inhibition of phosphatase binding.
Collapse
Affiliation(s)
- W Gordon-Krajcer
- Department of Pathology, Rm. F-538, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA
| | | | | |
Collapse
|
49
|
Buée L, Mailliot C, Bussière T, Sergeant N, Buée-Scherrer V, Hof PR, Flament S, Delacourte A. Neurodegenerative Disorders with Tauopathies: Mad Tau Diseases? FATAL ATTRACTIONS: PROTEIN AGGREGATES IN NEURODEGENERATIVE DISORDERS 2000. [DOI: 10.1007/978-3-662-04056-0_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
50
|
Buée L, Delacourte A. Comparative biochemistry of tau in progressive supranuclear palsy, corticobasal degeneration, FTDP-17 and Pick's disease. Brain Pathol 1999; 9:681-93. [PMID: 10517507 PMCID: PMC8098140 DOI: 10.1111/j.1750-3639.1999.tb00550.x] [Citation(s) in RCA: 259] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Neurodegenerative disorders referred to as tauopathies have cellular hyperphosphorylated tau protein aggregates in the absence of amyloid deposits. Comparative biochemistry of tau aggregates shows that they differ in both phosphorylation and content of tau isoforms. The six tau isoforms found in human brain contain either three (3R) or four microtubule-binding domains (4R). In Alzheimer's disease, all six tau isoforms are abnormally phosphorylated and aggregate into paired helical filaments. They are detected by immunoblotting as a major tau triplet (tau55, 64 and 69). In corticobasal degeneration and progressive supranuclear palsy, only 4R-tau isoforms aggregate into twisted and straight filaments respectively. They appear as a major tau doublet (tau64 and 69). Finally, in Pick's disease, only 3R-tau isoforms aggregate into random coiled filaments. They are characterized by another major tau doublet (tau55 and 64). These differences in tau isoforms may be related to either the degeneration of particular cell populations in a given disorder or aberrant cell trafficking of particular tau isoforms. Finally, recent findings provide a direct link between a genetic defect in tau and its abnormal aggregation into filaments in fronto-temporal dementia with Parkinsonism linked to chromosome 17, demonstrating that tau aggregation is sufficient for nerve cell degeneration. Thus, tau mutations and polymorphisms may also be instrumental in many neurodegenerative disorders.
Collapse
Affiliation(s)
- L Buée
- INSERM U422, F-59045 Lille, France.
| | | |
Collapse
|