1
|
Uliassi E, Bolognesi ML, Milelli A. Targeting Tau Protein with Proximity Inducing Modulators: A New Frontier to Combat Tauopathies. ACS Pharmacol Transl Sci 2025; 8:654-672. [PMID: 40109749 PMCID: PMC11915046 DOI: 10.1021/acsptsci.4c00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 03/22/2025]
Abstract
Dysregulation of correct protein tau homeostasis represents the seed for the development of several devastating central nervous system disorders, known as tauopathies, that affect millions of people worldwide. Despite massive public and private support to research funding, these diseases still represent unmet medical needs. In fact, the tau-targeting tools developed to date have failed to translate into the clinic. Recently, taking advantage of the modes that nature uses to mediate the flow of information in cells, researchers have developed a new class of molecules, called proximity-inducing modulators, which exploit spatial proximity to modulate protein function(s) and redirect cellular processes. In this perspective, after a brief discussion about tau protein and the classic tau-targeting approaches, we will discuss the different classes of proximity-inducing modulators developed so far and highlight the applications to modulate tau protein's function and tau-induced toxicity.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Andrea Milelli
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d'Augusto 237, Rimini 47921, Italy
| |
Collapse
|
2
|
Kuznetsov IA, Kuznetsov AV. Why slow axonal transport is bidirectional - can axonal transport of tau protein rely only on motor-driven anterograde transport? Comput Methods Biomech Biomed Engin 2024; 27:620-631. [PMID: 37068039 DOI: 10.1080/10255842.2023.2197541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/27/2023] [Indexed: 04/18/2023]
Abstract
Slow axonal transport (SAT) moves multiple proteins from the soma, where they are synthesized, to the axon terminal. Due to the great lengths of axons, SAT almost exclusively relies on active transport, which is driven by molecular motors. The puzzling feature of slow axonal transport is its bidirectionality. Although the net direction of SAT is anterograde, from the soma to the terminal, experiments show that it also contains a retrograde component. One of the proteins transported by SAT is the microtubule-associated protein tau. To better understand why the retrograde component in tau transport is needed, we used the perturbation technique to analyze how the full tau SAT model can be simplified for the specific case when retrograde motor-driven transport and diffusion-driven transport of tau are negligible and tau is driven only by anterograde (kinesin) motors. The solution of the simplified equations shows that without retrograde transport the tau concentration along the axon length stays almost uniform (decreases very slightly), which is inconsistent with the experimenal tau concentration at the outlet boundary (at the axon tip). Thus kinesin-driven transport alone is not enough to explain the empirically observed distribution of tau, and the retrograde motor-driven component in SAT is needed.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
3
|
Hoshino A, Clemente V, Shetty M, Castle B, Odde D, Bazzaro M. The microtubule-severing protein UNC-45A preferentially binds to curved microtubules and counteracts the microtubule-straightening effects of Taxol. J Biol Chem 2023; 299:105355. [PMID: 37858676 PMCID: PMC10654038 DOI: 10.1016/j.jbc.2023.105355] [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: 02/22/2023] [Revised: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023] Open
Abstract
Uncoordinated protein 45A (UNC-45A) is the only known ATP-independent microtubule (MT)-severing protein. Thus, it severs MTs via a novel mechanism. In vitro and in cells, UNC-45A-mediated MT severing is preceded by the appearance of MT bends. While MTs are stiff biological polymers, in cells, they often curve, and the result of this curving can be breaking off. The contribution of MT-severing proteins on MT lattice curvature is largely undefined. Here, we show that UNC-45A curves MTs. Using in vitro biophysical reconstitution and total internal fluorescence microscopy analysis, we show that UNC-45A is enriched in the areas where MTs are curved versus the areas where MTs are straight. In cells, we show that UNC-45A overexpression increases MT curvature and its depletion has the opposite effect. We also show that this effect occurs is independent of actomyosin contractility. Lastly, we show for the first time that in cells, Paclitaxel straightens MTs, and that UNC-45A can counteracts the MT-straightening effects of the drug.
Collapse
Affiliation(s)
- Asumi Hoshino
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Valentino Clemente
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mihir Shetty
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brian Castle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - David Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
4
|
Tu Z, Yan S, Han B, Li C, Liang W, Lin Y, Ding Y, Wei H, Wang L, Xu H, Ye J, Li B, Li S, Li XJ. Tauopathy promotes spinal cord-dependent production of toxic amyloid-beta in transgenic monkeys. Signal Transduct Target Ther 2023; 8:358. [PMID: 37735155 PMCID: PMC10514290 DOI: 10.1038/s41392-023-01601-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 09/23/2023] Open
Abstract
Tauopathy, characterized by the hyperphosphorylation and accumulation of the microtubule-associated protein tau, and the accumulation of Aβ oligomers, constitute the major pathological hallmarks of Alzheimer's disease. However, the relationship and causal roles of these two pathological changes in neurodegeneration remain to be defined, even though they occur together or independently in several neurodegenerative diseases associated with cognitive and movement impairment. While it is widely accepted that Aβ accumulation leads to tauopathy in the late stages of the disease, it is still unknown whether tauopathy influences the formation of toxic Aβ oligomers. To address this, we generated transgenic cynomolgus monkey models expressing Tau (P301L) through lentiviral infection of monkey embryos. These monkeys developed age-dependent neurodegeneration and motor dysfunction. Additionally, we performed a stereotaxic injection of adult monkey and mouse brains to express Tau (P301L) via AAV9 infection. Importantly, we found that tauopathy resulting from embryonic transgenic Tau expression or stereotaxic brain injection of AAV-Tau selectively promoted the generation of Aβ oligomers in the monkey spinal cord. These Aβ oligomers were recognized by several antibodies to Aβ1-42 and contributed to neurodegeneration. However, the generation of Aβ oligomers was not observed in other brain regions of Tau transgenic monkeys or in the brains of mice injected with AAV9-Tau (P301L), suggesting that the generation of Aβ oligomers is species- and brain region-dependent. Our findings demonstrate for the first time that tauopathy can trigger Aβ pathology in the primate spinal cord and provide new insight into the pathogenesis and treatment of tauopathy.
Collapse
Affiliation(s)
- Zhuchi Tu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Sen Yan
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Bofeng Han
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Caijuan Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Weien Liang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yingqi Lin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yongyan Ding
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jianmeng Ye
- Guangdong Landau Biotechnology Co. Ltd., Guangzhou, 510555, China
| | - Bang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
5
|
Hoshino A, Clemente V, Shetty M, Castle B, Odde D, Bazzaro M. The Microtubule Severing Protein UNC-45A Counteracts the Microtubule Straightening Effects of Taxol. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557417. [PMID: 37745537 PMCID: PMC10515786 DOI: 10.1101/2023.09.12.557417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
UNC-45A is the only known ATP-independent microtubule (MT) severing protein. Thus, it severs MTs via a novel mechanism. In vitro and in cells UNC-45A-mediated MT severing is preceded by the appearance of MT bends. While MTs are stiff biological polymers, in cells, they often curve, and the result of this curving can be breaking off. The contribution of MT severing proteins on MT lattice curvature is largely undefined. Here we show that UNC-45A curves MTs. Using in vitro biophysical reconstitution and TIRF microscopy analysis, we show that UNC-45A is enriched in the areas where MTs are curved versus the areas where MTs are straight. In cells, we show that UNC-45A overexpression increases MT curvature and its depletion has the opposite effect. We also show that this effect occurs is independent of actomyosin contractility. Lastly, we show for the first time that in cells, Paclitaxel straightens MTs, and that UNC-45A can counteracts the MT straightening effects of the drug. Significance: Our findings reveal for the first time that UNC-45A increases MT curvature. This hints that UNC-45A-mediated MT severing could be due to the worsening of MT curvature and provide a mechanistic understanding of how this MT-severing protein may act. UNC-45A is the only MT severing protein expressed in human cancers, including paclitaxel-resistant ovarian cancer. Our finding that UNC-45A counteracts the paclitaxel-straightening effects of MTs in cells suggests an additional mechanism through which cancer cells escape drug treatment.
Collapse
|
6
|
Limorenko G, Tatli M, Kolla R, Nazarov S, Weil MT, Schöndorf DC, Geist D, Reinhardt P, Ehrnhoefer DE, Stahlberg H, Gasparini L, Lashuel HA. Fully co-factor-free ClearTau platform produces seeding-competent Tau fibrils for reconstructing pathological Tau aggregates. Nat Commun 2023; 14:3939. [PMID: 37402718 PMCID: PMC10319797 DOI: 10.1038/s41467-023-39314-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/06/2023] [Indexed: 07/06/2023] Open
Abstract
Tau protein fibrillization is implicated in the pathogenesis of several neurodegenerative diseases collectively known as Tauopathies. For decades, investigating Tau fibrillization in vitro has required the addition of polyanions or other co-factors to induce its misfolding and aggregation, with heparin being the most commonly used. However, heparin-induced Tau fibrils exhibit high morphological heterogeneity and a striking structural divergence from Tau fibrils isolated from Tauopathies patients' brains at ultra- and macro-structural levels. To address these limitations, we developed a quick, cheap, and effective method for producing completely co-factor-free fibrils from all full-length Tau isoforms and mixtures thereof. We show that Tau fibrils generated using this ClearTau method - ClearTau fibrils - exhibit amyloid-like features, possess seeding activity in biosensor cells and hiPSC-derived neurons, retain RNA-binding capacity, and have morphological properties and structures more reminiscent of the properties of the brain-derived Tau fibrils. We present the proof-of-concept implementation of the ClearTau platform for screening Tau aggregation-modifying compounds. We demonstrate that these advances open opportunities to investigate the pathophysiology of disease-relevant Tau aggregates and will facilitate the development of Tau pathology-targeting and modifying therapies and PET tracers that can distinguish between different Tauopathies.
Collapse
Affiliation(s)
- Galina Limorenko
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Meltem Tatli
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Rajasekhar Kolla
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Sergey Nazarov
- Biological Electron Microscopy Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Marie-Theres Weil
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061, Ludwigshafen, Germany
| | - David C Schöndorf
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Daniela Geist
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Peter Reinhardt
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Dagmar E Ehrnhoefer
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Henning Stahlberg
- Laboratory of Biological Electron Microscopy, Institute of Physics, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
- Department of Fund. Microbiology, Faculty of Biology and Medicine, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Laura Gasparini
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland.
| |
Collapse
|
7
|
Balabanian L, Lessard DV, Swaminathan K, Yaninska P, Sébastien M, Wang S, Stevens PW, Wiseman PW, Berger CL, Hendricks AG. Tau differentially regulates the transport of early endosomes and lysosomes. Mol Biol Cell 2022; 33:ar128. [PMID: 36129768 PMCID: PMC9634973 DOI: 10.1091/mbc.e22-01-0018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Microtubule-associated proteins (MAPs) modulate the motility of kinesin and dynein along microtubules to control the transport of vesicles and organelles. The neuronal MAP tau inhibits kinesin-dependent transport. Phosphorylation of tau at Tyr-18 by fyn kinase results in weakened inhibition of kinesin-1. We examined the motility of early endosomes and lysosomes in cells expressing wild-type (WT) tau and phosphomimetic Y18E tau. We quantified the effects on motility as a function of the tau expression level. Lysosome motility is strongly inhibited by tau. Y18E tau preferentially inhibits lysosomes in the cell periphery, while centrally located lysosomes are less affected. Early endosomes are more sensitive to tau than lysosomes and are inhibited by both WT and Y18E tau. Our results show that different cargoes have disparate responses to tau, likely governed by the types of kinesin motors driving their transport. In support of this model, kinesin-1 and -3 are strongly inhibited by tau while kinesin-2 and dynein are less affected. In contrast to kinesin-1, we find that kinesin-3 is strongly inhibited by phosphorylated tau.
Collapse
Affiliation(s)
- Linda Balabanian
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Dominique V. Lessard
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405
| | | | - Pamela Yaninska
- Chemistry and Physics, McGill University, Montreal, QC H3A 0E9, Canada
| | - Muriel Sébastien
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Samuel Wang
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Piper W. Stevens
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Paul W. Wiseman
- Chemistry and Physics, McGill University, Montreal, QC H3A 0E9, Canada
| | - Christopher L. Berger
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405
| | - Adam G. Hendricks
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada,*Address correspondence to: Adam G. Hendricks ()
| |
Collapse
|
8
|
Siahaan V, Tan R, Humhalova T, Libusova L, Lacey SE, Tan T, Dacy M, Ori-McKenney KM, McKenney RJ, Braun M, Lansky Z. Microtubule lattice spacing governs cohesive envelope formation of tau family proteins. Nat Chem Biol 2022; 18:1224-1235. [PMID: 35996000 PMCID: PMC9613621 DOI: 10.1038/s41589-022-01096-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/24/2022] [Indexed: 01/28/2023]
Abstract
Tau is an intrinsically disordered microtubule-associated protein (MAP) implicated in neurodegenerative disease. On microtubules, tau molecules segregate into two kinetically distinct phases, consisting of either independently diffusing molecules or interacting molecules that form cohesive 'envelopes' around microtubules. Envelopes differentially regulate lattice accessibility for other MAPs, but the mechanism of envelope formation remains unclear. Here we find that tau envelopes form cooperatively, locally altering the spacing of tubulin dimers within the microtubule lattice. Envelope formation compacted the underlying lattice, whereas lattice extension induced tau envelope disassembly. Investigating other members of the tau family, we find that MAP2 similarly forms envelopes governed by lattice spacing, whereas MAP4 cannot. Envelopes differentially biased motor protein movement, suggesting that tau family members could spatially divide the microtubule surface into functionally distinct regions. We conclude that the interdependent allostery between lattice spacing and cooperative envelope formation provides the molecular basis for spatial regulation of microtubule-based processes by tau and MAP2.
Collapse
Affiliation(s)
- Valerie Siahaan
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prague West, Czech Republic
| | - Ruensern Tan
- Department of Molecular and Cellular Biology, University of California - Davis, Davis, CA, USA
- Department of Molecular and Cellular Biology, University of California at Berkeley, Berkeley, CA, USA
| | - Tereza Humhalova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Lenka Libusova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Samuel E Lacey
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, UK
- Human Technopole, Milan, Italy
| | - Tracy Tan
- Department of Molecular and Cellular Biology, University of California - Davis, Davis, CA, USA
| | - Mariah Dacy
- Department of Molecular and Cellular Biology, University of California - Davis, Davis, CA, USA
| | | | - Richard J McKenney
- Department of Molecular and Cellular Biology, University of California - Davis, Davis, CA, USA.
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prague West, Czech Republic.
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prague West, Czech Republic.
| |
Collapse
|
9
|
Fourest-Lieuvin A, Vinit A, Blot B, Perrot A, Denarier E, Saudou F, Arnal I. Controlled Tau Cleavage in Cells Reveals Abnormal Localizations of Tau Fragments. Neuroscience 2022; 518:162-177. [PMID: 35995336 DOI: 10.1016/j.neuroscience.2022.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/08/2022] [Accepted: 08/16/2022] [Indexed: 11/15/2022]
Abstract
In several forms of dementia, such as Alzheimer's disease, the cytoskeleton-associated protein tau undergoes proteolysis, giving rise to fragments that have a toxic impact on neuronal homeostasis. How these fragments interact with cellular structures, in particular with the cytoskeleton, is currently incompletely understood. Here, we developed a method, derived from a Tobacco Etch Virus (TEV) protease system, to induce controlled cleavage of tau at specific sites. Five tau proteins containing specific TEV recognition sites corresponding to pathological proteolytic sites were engineered, and tagged with GFP at one end and mCherry at the other. Following controlled cleavage to produce GFP-N-terminal and C-terminal-mCherry fragments, we followed the fate of tau fragments in cells. Our results showed that whole engineered tau proteins associate with the cytoskeleton similarly to the non-modified tau, whereas tau fragments adopted different localizations with respect to the actin and microtubule cytoskeletons. These distinct localizations were confirmed by expressing each separate fragment in cells. Some cleavages - in particular cleavages at amino-acid positions 124 or 256 - displayed a certain level of cellular toxicity, with an unusual relocalization of the N-terminal fragments to the nucleus. Based on the data presented here, inducible cleavage of tau by the TEV protease appears to be a valuable tool to reproduce tau fragmentation in cells and study the resulting consequences on cell physiology.
Collapse
Affiliation(s)
- Anne Fourest-Lieuvin
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| | - Angélique Vinit
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Béatrice Blot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Anthime Perrot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eric Denarier
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frédéric Saudou
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Isabelle Arnal
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
10
|
Jackson NA, Guerrero-Muñoz MJ, Castillo-Carranza DL. The prion-like transmission of tau oligomers via exosomes. Front Aging Neurosci 2022; 14:974414. [PMID: 36062141 PMCID: PMC9434014 DOI: 10.3389/fnagi.2022.974414] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
The conversion and transmission of misfolded proteins established the basis for the prion concept. Neurodegenerative diseases are considered “prion-like” disorders that lack infectivity. Among them, tauopathies are characterized by the conversion of native tau protein into an abnormally folded aggregate. During the progression of the disease, misfolded tau polymerizes into oligomers and intracellular neurofibrillary tangles (NFTs). While the toxicity of NFTs is an ongoing debate, the contribution of tau oligomers to early onset neurodegenerative pathogenesis is accepted. Tau oligomers are readily transferred from neuron to neuron propagating through the brain inducing neurodegeneration. Recently, transmission of tau oligomers via exosomes is now proposed. There is still too much to uncover about tau misfolding and propagation. Here we summarize novel findings of tau oligomers transmission and propagation via exosomes.
Collapse
Affiliation(s)
- Noel A. Jackson
- School of Public Health, Harvard University, Boston, MA, United States
| | | | - Diana L. Castillo-Carranza
- School of Medicine, University of Monterrey, San Pedro Garza García, Mexico
- *Correspondence: Diana L. Castillo-Carranza,
| |
Collapse
|
11
|
Kulkarni R, Thakur A, Kumar H. Microtubule Dynamics Following Central and Peripheral Nervous System Axotomy. ACS Chem Neurosci 2022; 13:1358-1369. [PMID: 35451811 DOI: 10.1021/acschemneuro.2c00189] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Disturbance in the neuronal network leads to instability in the microtubule (MT) railroad of axons, causing hindrance in the intra-axonal transport and making it difficult to re-establish the broken network. Peripheral nervous system (PNS) neurons can stabilize their MTs, leading to the formation of regeneration-promoting structures called "growth cones". However, central nervous system (CNS) neurons lack this intrinsic reparative capability and, instead, form growth-incompetent structures called "retraction bulbs", which have a disarrayed MT network. It is evident from various studies that although axonal regeneration depends on both cell-extrinsic and cell-intrinsic factors, any therapy that aims at axonal regeneration ultimately converges onto MTs. Understanding the neuronal MT dynamics will help develop effective therapeutic strategies in diseases where the MT network gets disrupted, such as spinal cord injury, traumatic brain injury, multiple sclerosis, and amyotrophic lateral sclerosis. It is also essential to know the factors that aid or inhibit MT stabilization. In this review, we have discussed the MT dynamics postaxotomy in the CNS and PNS, and factors that can directly influence MT stability in various diseases.
Collapse
Affiliation(s)
- Riya Kulkarni
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Akshata Thakur
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
12
|
Soliman A, Bakota L, Brandt R. Microtubule-modulating Agents in the Fight Against Neurodegeneration: Will it ever Work? Curr Neuropharmacol 2022; 20:782-798. [PMID: 34852744 PMCID: PMC9878958 DOI: 10.2174/1570159x19666211201101020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022] Open
Abstract
The microtubule skeleton plays an essential role in nerve cells as the most important structural determinant of morphology and as a highway for axonal transport processes. Many neurodegenerative diseases are characterized by changes in the structure and organization of microtubules and microtubule-regulating proteins such as the microtubule-associated protein tau, which exhibits characteristic changes in a whole class of diseases collectively referred to as tauopathies. Changes in the dynamics of microtubules appear to occur early under neurodegenerative conditions and are also likely to contribute to age-related dysfunction of neurons. Thus, modulating microtubule dynamics and correcting impaired microtubule stability can be a useful neuroprotective strategy to counteract the disruption of the microtubule system in disease and aging. In this article, we review current microtubule- directed approaches for the treatment of neurodegenerative diseases with microtubules as a drug target, tau as a drug target, and post-translational modifications as potential modifiers of the microtubule system. We discuss limitations of the approaches that can be traced back to the rather unspecific mechanism of action, which causes undesirable side effects in non-neuronal cell types or which are due to the disruption of non-microtubule-related interactions. We also develop some thoughts on how the specificity of the approaches can be improved and what further targets could be used for modulating substances.
Collapse
Affiliation(s)
- Ahmed Soliman
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany;,Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany;,Institute of Cognitive Science, Osnabrück University, Osnabrück, Germany,Address correspondence to this author at the Department of Neurobiology, Osnabrück University, Osnabrück, Germany; Tel: +49 541 969 2338; E-mail:
| |
Collapse
|
13
|
Limorenko G, Lashuel HA. Revisiting the grammar of Tau aggregation and pathology formation: how new insights from brain pathology are shaping how we study and target Tauopathies. Chem Soc Rev 2021; 51:513-565. [PMID: 34889934 DOI: 10.1039/d1cs00127b] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Converging evidence continues to point towards Tau aggregation and pathology formation as central events in the pathogenesis of Alzheimer's disease and other Tauopathies. Despite significant advances in understanding the morphological and structural properties of Tau fibrils, many fundamental questions remain about what causes Tau to aggregate in the first place. The exact roles of cofactors, Tau post-translational modifications, and Tau interactome in regulating Tau aggregation, pathology formation, and toxicity remain unknown. Recent studies have put the spotlight on the wide gap between the complexity of Tau structures, aggregation, and pathology formation in the brain and the simplicity of experimental approaches used for modeling these processes in research laboratories. Embracing and deconstructing this complexity is an essential first step to understanding the role of Tau in health and disease. To help deconstruct this complexity and understand its implication for the development of effective Tau targeting diagnostics and therapies, we firstly review how our understanding of Tau aggregation and pathology formation has evolved over the past few decades. Secondly, we present an analysis of new findings and insights from recent studies illustrating the biochemical, structural, and functional heterogeneity of Tau aggregates. Thirdly, we discuss the importance of adopting new experimental approaches that embrace the complexity of Tau aggregation and pathology as an important first step towards developing mechanism- and structure-based therapies that account for the pathological and clinical heterogeneity of Alzheimer's disease and Tauopathies. We believe that this is essential to develop effective diagnostics and therapies to treat these devastating diseases.
Collapse
Affiliation(s)
- Galina Limorenko
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
14
|
Khan MI, Gilpin K, Hasan F, Mahmud KAHA, Adnan A. Effect of Strain Rate on Single Tau, Dimerized Tau and Tau-Microtubule Interface: A Molecular Dynamics Simulation Study. Biomolecules 2021; 11:1308. [PMID: 34572521 PMCID: PMC8472149 DOI: 10.3390/biom11091308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 01/24/2023] Open
Abstract
Microtubule-associated protein (MAP) tau is a cross-linking molecule that provides structural stability to axonal microtubules (MT). It is considered a potential biomarker for Alzheimer's disease (AD), dementia, and other neurological disorders. It is also a signature protein for Traumatic Brain Injury (TBI) assessment. In the case of TBI, extreme dynamic mechanical energies can be felt by the axonal cytoskeletal members. As such, fundamental understandings of the responses of single tau protein, polymerized tau protein, and tau-microtubule interfaces under high-rate mechanical forces are important. This study attempts to determine the high-strain rate mechanical behavior of single tau, dimerized tau, and tau-MT interface using molecular dynamics (MD) simulation. The results show that a single tau protein is a highly stretchable soft polymer. During deformation, first, it significantly unfolds against van der Waals and electrostatic bonds. Then it stretches against strong covalent bonds. We found that tau acts as a viscoelastic material, and its stiffness increases with the strain rate. The unfolding stiffness can be ~50-500 MPa, while pure stretching stiffness can be >2 GPa. The dimerized tau model exhibits similar behavior under similar strain rates, and tau sliding from another tau is not observed until it is stretched to >7 times of original length, depending on the strain rate. The tau-MT interface simulations show that very high strain and strain rates are required to separate tau from MT suggesting Tau-MT bonding is stronger than MT subunit bonding between themselves. The dimerized tau-MT interface simulations suggest that tau-tau bonding is stronger than tau-MT bonding. In summary, this study focuses on the structural response of individual cytoskeletal components, namely microtubule (MT) and tau protein. Furthermore, we consider not only the individual response of a component, but also their interaction with each other (such as tau with tau or tau with MT). This study will eventually pave the way to build a bottom-up multiscale brain model and analyze TBI more comprehensively.
Collapse
Affiliation(s)
- Md Ishak Khan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| | - Kathleen Gilpin
- Academic Partnership and Engagement Experiment (APEX), Wright State Applied Research Corporation, Beavercreek, OH 45431, USA;
| | - Fuad Hasan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| | - Khandakar Abu Hasan Al Mahmud
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| | - Ashfaq Adnan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019, USA; (M.I.K.); (F.H.); (K.A.H.A.M.)
| |
Collapse
|
15
|
Castle BT, McKibben KM, Rhoades E, Odde DJ. Tau Avoids the GTP Cap at Growing Microtubule Plus-Ends. iScience 2020; 23:101782. [PMID: 33294790 PMCID: PMC7691178 DOI: 10.1016/j.isci.2020.101782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/15/2020] [Accepted: 11/04/2020] [Indexed: 10/26/2022] Open
Abstract
Plus-end tracking proteins (+TIPs) associate with the growing end of microtubules and mediate important cellular functions. The majority of +TIPs are directed to the plus-end through a family of end-binding proteins (EBs), which preferentially bind the stabilizing cap of GTP-tubulin present during microtubule growth. One outstanding question is whether there may exist other microtubule-associated proteins (MAPs) that preferentially bind specific nucleotide states of tubulin. Here, we report that the neuronal MAP tau preferentially binds GDP-tubulin (K D = 0.26 μM) over GMPCPP-tubulin (K D = 1.1 μM) in vitro, as well as GTP-tubulin at the tips of growing microtubules, causing tau binding to lag behind the plus-end both in vitro and in live cells. Thus, tau is a microtubule tip avoiding protein, establishing the framework for a possible new class of tip avoiding MAPs. We speculate that disease-relevant tau mutations may exert their phenotype by their failure to properly recognize GDP-tubulin.
Collapse
Affiliation(s)
- Brian T. Castle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kristen M. McKibben
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth Rhoades
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J. Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
16
|
Alyenbaawi H, Allison WT, Mok SA. Prion-Like Propagation Mechanisms in Tauopathies and Traumatic Brain Injury: Challenges and Prospects. Biomolecules 2020; 10:E1487. [PMID: 33121065 PMCID: PMC7692808 DOI: 10.3390/biom10111487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022] Open
Abstract
The accumulation of tau protein in the form of filamentous aggregates is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). These dementias share traumatic brain injury (TBI) as a prominent risk factor. Tau aggregates can transfer between cells and tissues in a "prion-like" manner, where they initiate the templated misfolding of normal tau molecules. This enables the spread of tau pathology to distinct parts of the brain. The evidence that tauopathies spread via prion-like mechanisms is considerable, but work detailing the mechanisms of spread has mostly used in vitro platforms that cannot fully reveal the tissue-level vectors or etiology of progression. We review these issues and then briefly use TBI and CTE as a case study to illustrate aspects of tauopathy that warrant further attention in vivo. These include seizures and sleep/wake disturbances, emphasizing the urgent need for improved animal models. Dissecting these mechanisms of tauopathy progression continues to provide fresh inspiration for the design of diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hadeel Alyenbaawi
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Medical Laboratories, Majmaah University, Majmaah 11952, Saudi Arabia
| | - W. Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Sue-Ann Mok
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
17
|
Khan MI, Hasan F, Hasan Al Mahmud KA, Adnan A. Domain focused and residue focused phosphorylation effect on tau protein: A molecular dynamics simulation study. J Mech Behav Biomed Mater 2020; 113:104149. [PMID: 33125954 DOI: 10.1016/j.jmbbm.2020.104149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 11/19/2022]
Abstract
Phosphorylation has been hypothesized to alter the ability of tau protein to bind with microtubules (MT), and pathological level of phosphorylation can incorporate formation of Paired Helical Filaments (PHF) in affected tau. Study of the effect of phosphorylation on different domains of tau (projection domain, microtubule binding sites and N-terminus tail) is important to obtain insight about tau neuropathology. In an earlier study, we have already obtained the mechanical properties and behavior of single tau and dimerized tau and observed tau-MT interaction for normal level of phosphorylation. This study attempts to obtain insights on the effect of phosphorylation on different domains of tau, using molecular dynamics (MD) simulation with the aid of CHARMM force field under high strain rate. It also determines the effect of residue focused phosphorylation on tau-MT interaction and tau accumulation tendency. The results show that for single tau protein, unfolding stiffness does not differ significantly due to phosphorylation, but stretching stiffness can be much higher than the normally phosphorylated protein. For dimerized tau protein, the stretching required to separate the protein forming the dimer is similar for phosphorylation in individual domains but is significantly less in case of phosphorylation in all domains. For tau-MT interaction simulations, it is found that for normal phosphorylation, the tau separation from MT occurs at higher strain for phosphorylation in projection domain and N-terminus tail, and earlier for phosphorylation in all domains altogether than the normal phosphorylation state. The residue focused phosphorylation study also shows that tau separates earlier from MT and shows stronger accumulation tendency at the phosphorylated state, while preserving the highly stretchable and flexible characteristic of tau. This study provides important insight on mechanochemical phenomena relevant to traumatic brain injury (TBI) scenario, where the result of mechanical loading and posttranslational modification as well as conformation decides the mechanical behavior.
Collapse
Affiliation(s)
- Md Ishak Khan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Fuad Hasan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | | | - Ashfaq Adnan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA.
| |
Collapse
|
18
|
Kuznetsov IA, Kuznetsov AV. Modeling tau transport in the axon initial segment. Math Biosci 2020; 329:108468. [PMID: 32920097 DOI: 10.1016/j.mbs.2020.108468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 11/18/2022]
Abstract
By assuming that tau protein can be in seven kinetic states, we developed a model of tau protein transport in the axon and in the axon initial segment (AIS). Two separate sets of kinetic constants were determined, one in the axon and the other in the AIS. This was done by fitting the model predictions in the axon with experimental results and by fitting the model predictions in the AIS with the assumed linear increase of the total tau concentration in the AIS. The calibrated model was used to make predictions about tau transport in the axon and in the AIS. To the best of our knowledge, this is the first paper that presents a mathematical model of tau transport in the AIS. Our modeling results suggest that binding of free tau to microtubules creates a negative gradient of free tau in the AIS. This leads to diffusion-driven tau transport from the soma into the AIS. The model further suggests that slow axonal transport and diffusion-driven transport of tau work together in the AIS, moving tau anterogradely. Our numerical results predict an interplay between these two mechanisms: as the distance from the soma increases, the diffusion-driven transport decreases, while motor-driven transport becomes larger. Thus, the machinery in the AIS works as a pump, moving tau into the axon.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
19
|
Li J, Li Y, Liu M, Xie S. Modified heptapeptide from tau binds both tubulin and microtubules. Thorac Cancer 2020; 11:2993-2997. [PMID: 32893987 PMCID: PMC7529580 DOI: 10.1111/1759-7714.13643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Background Microtubules are the major cytoskeletal component in eukaryotes which are essential for a large spectrum of cellular activities. Monitoring the behavior of microtubules is helpful for a better understanding of the regulatory mechanism governing microtubule architecture and microtubule‐based activities. Here, we characterized the binding capability of a modified heptapeptide from tau to both tubulin and microtubules and sought to develop it as a fluorescent peptide for monitoring microtubules. Methods To deliver the fluorescent peptide into the cells, a cell‐penetrating peptide was conjugated to the modified heptapeptide from tau and synthesized. The affinity of the modified heptapeptide was determined by microscale thermophoresis. The microtubule labeling ability was determined by adding the peptide into the polymerized microtubule solutions or cultured HeLa cells.; Results Affinity determination revealed that the tau‐derived peptide specifically bound to tubulin. In addition, the peptide was able to label polymerized microtubules in solution, although no obvious microtubule filaments were observed clearly in living cells, probably due to the inadequate affinity. Conclusions These results suggest that using a peptide‐based strategy for imaging microtubules might be plausible and attempts to improve its affinity is warranted in the future.
Collapse
Affiliation(s)
- Jingrui Li
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Yuyang Li
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Min Liu
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Songbo Xie
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
20
|
Prokop A. Cytoskeletal organization of axons in vertebrates and invertebrates. J Cell Biol 2020; 219:e201912081. [PMID: 32369543 PMCID: PMC7337489 DOI: 10.1083/jcb.201912081] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
The maintenance of axons for the lifetime of an organism requires an axonal cytoskeleton that is robust but also flexible to adapt to mechanical challenges and to support plastic changes of axon morphology. Furthermore, cytoskeletal organization has to adapt to axons of dramatically different dimensions, and to their compartment-specific requirements in the axon initial segment, in the axon shaft, at synapses or in growth cones. To understand how the cytoskeleton caters to these different demands, this review summarizes five decades of electron microscopic studies. It focuses on the organization of microtubules and neurofilaments in axon shafts in both vertebrate and invertebrate neurons, as well as the axon initial segments of vertebrate motor- and interneurons. Findings from these ultrastructural studies are being interpreted here on the basis of our contemporary molecular understanding. They strongly suggest that axon architecture in animals as diverse as arthropods and vertebrates is dependent on loosely cross-linked bundles of microtubules running all along axons, with only minor roles played by neurofilaments.
Collapse
Affiliation(s)
- Andreas Prokop
- School of Biology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Ahmad W, Shabbiri K, Ahmad I. Prediction of human tau 3D structure, and interplay between O-β-GlcNAc and phosphorylation modifications in Alzheimer's disease: C. elegans as a suitable model to study these interactions in vivo. Biochem Biophys Res Commun 2020; 528:466-472. [PMID: 32499112 DOI: 10.1016/j.bbrc.2020.05.176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022]
Abstract
Tau protein regulates, maintains and stabilizes microtubule assembly under normal physiological conditions. In certain pathological circumstances, tau is post-translationally modified predominantly via phosphorylation and glycosylation. Hyper-phosphorylation of tau in Alzheimer's disease (AD) resulted in aggregated neurofibrillary tangles (NFTs) formation. Unfortunately, absence of tau 3D structure makes difficult to understand exact mechanism involved in tau pathology. Here by using ab-initio modelling, we predicted a tau 3D structure that not only explains its binding with microtubules but also elucidates NFTs formation. O-linked β-N-acetylglucosaminylation (O-β-GlcNAc) is thought to regulate tau phosphorylation on single or proximal Ser/Thr residues (called as Yin-Yang sites). In this study, we not only validate the previously described three-serine residues (208, 238 and 400) as Yin-Yang sites but also predicted 22 more possible Ser/Thr O-glycosylation sites. Among them seventeen residues were predicted as possible Yin-Yang sites and are proposed to mediate NFT formation in AD. These predicted Yin-Yang sites may act as attractive therapeutic targets for the drug development in AD. Predicted 3D structure of tau441 was highly accessible for phosphorylation and hyperphosphorylation, and showed higher surface accessibility for interplay between O-β-GlcNAc and phosphorylation modifications. Kinases and phosphatases involved in tau phosphorylation are conserved in human and other organisms. Homology modelling revealed conserved catalytic domain for both human and C. elegans O-GlcNAc transferase (OGT), suggesting that transgenic C. elegans expressing human tau may be a suitable model system to study these modifications.
Collapse
Affiliation(s)
- Waqar Ahmad
- School of Biological Sciences, The University of Queensland, Brisbane, 4072, Australia.
| | - Khadija Shabbiri
- School of Biological Sciences, The University of Queensland, Brisbane, 4072, Australia.
| | - Ishtiaq Ahmad
- Department of Chemistry, GC University, Lahore, 54000, Pakistan.
| |
Collapse
|
22
|
Hahn I, Voelzmann A, Liew YT, Costa-Gomes B, Prokop A. The model of local axon homeostasis - explaining the role and regulation of microtubule bundles in axon maintenance and pathology. Neural Dev 2019; 14:11. [PMID: 31706327 PMCID: PMC6842214 DOI: 10.1186/s13064-019-0134-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Axons are the slender, cable-like, up to meter-long projections of neurons that electrically wire our brains and bodies. In spite of their challenging morphology, they usually need to be maintained for an organism's lifetime. This makes them key lesion sites in pathological processes of ageing, injury and neurodegeneration. The morphology and physiology of axons crucially depends on the parallel bundles of microtubules (MTs), running all along to serve as their structural backbones and highways for life-sustaining cargo transport and organelle dynamics. Understanding how these bundles are formed and then maintained will provide important explanations for axon biology and pathology. Currently, much is known about MTs and the proteins that bind and regulate them, but very little about how these factors functionally integrate to regulate axon biology. As an attempt to bridge between molecular mechanisms and their cellular relevance, we explain here the model of local axon homeostasis, based on our own experiments in Drosophila and published data primarily from vertebrates/mammals as well as C. elegans. The model proposes that (1) the physical forces imposed by motor protein-driven transport and dynamics in the confined axonal space, are a life-sustaining necessity, but pose a strong bias for MT bundles to become disorganised. (2) To counterbalance this risk, MT-binding and -regulating proteins of different classes work together to maintain and protect MT bundles as necessary transport highways. Loss of balance between these two fundamental processes can explain the development of axonopathies, in particular those linking to MT-regulating proteins, motors and transport defects. With this perspective in mind, we hope that more researchers incorporate MTs into their work, thus enhancing our chances of deciphering the complex regulatory networks that underpin axon biology and pathology.
Collapse
Affiliation(s)
- Ines Hahn
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - André Voelzmann
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Yu-Ting Liew
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Beatriz Costa-Gomes
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Andreas Prokop
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK.
| |
Collapse
|
23
|
Kinetically distinct phases of tau on microtubules regulate kinesin motors and severing enzymes. Nat Cell Biol 2019; 21:1086-1092. [DOI: 10.1038/s41556-019-0374-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 07/18/2019] [Indexed: 11/09/2022]
|
24
|
Ukmar-Godec T, Wegmann S, Zweckstetter M. Biomolecular condensation of the microtubule-associated protein tau. Semin Cell Dev Biol 2019; 99:202-214. [PMID: 31260737 DOI: 10.1016/j.semcdb.2019.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
Cells contain multiple compartments dedicated to the regulation and control of biochemical reactions. Cellular compartments that are not surrounded by membranes can rapidly form and dissolve in response to changes in the cellular environment. The physicochemical processes that underlie the formation of non-membrane-bound compartments in vivo are connected to liquid-liquid phase separation of proteins and nucleic acids in vitro. Recent evidence suggests that the protein tau, which plays an important role in Alzheimer's disease and other neurodegenerative disorders, phase separates in solution, forms tau phases with microtubules, and associates with phase-separated RNA-binding protein granules in cells. Here we review the experimental evidence that supports the ability of tau to phase separate in solution and form biomolecular condensates in cells. As for other disease-relevant proteins, the physiological and pathological functions of tau are tightly connected - through loss of normal function or gain of toxic function - and we therefore discuss how tau phase separation plays a role for both, and with respect to different cellular functions of tau.
Collapse
Affiliation(s)
- Tina Ukmar-Godec
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany; Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Chariteplatz 1, 10117, Berlin, Germany.
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany; Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany; Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
25
|
Best RL, LaPointe NE, Liang J, Ruan K, Shade MF, Wilson L, Feinstein SC. Tau isoform-specific stabilization of intermediate states during microtubule assembly and disassembly. J Biol Chem 2019; 294:12265-12280. [PMID: 31266806 DOI: 10.1074/jbc.ra119.009124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/24/2019] [Indexed: 01/27/2023] Open
Abstract
The microtubule (MT)-associated protein tau regulates the critical growing and shortening behaviors of MTs, and its normal activity is essential for neuronal development and maintenance. Accordingly, aberrant tau action is tightly associated with Alzheimer's disease and is genetically linked to several additional neurodegenerative diseases known as tauopathies. Although tau is known to promote net MT growth and stability, the precise mechanistic details governing its regulation of MT dynamics remain unclear. Here, we have used the slowly-hydrolyzable GTP analog, guanylyl-(α,β)-methylene-diphosphonate (GMPCPP), to examine the structural effects of tau at MT ends that may otherwise be too transient to observe. The addition of both four-repeat (4R) and three-repeat (3R) tau isoforms to pre-formed GMPCPP MTs resulted in the formation of extended, multiprotofilament-wide projections at MT ends. Furthermore, at temperatures too low for assembly of bona fide MTs, both tau isoforms promoted the formation of long spiral ribbons from GMPCPP tubulin heterodimers. In addition, GMPCPP MTs undergoing cold-induced disassembly in the presence of 4R tau (and to a much lesser extent 3R tau) also formed spirals. Finally, three pathological tau mutations known to cause neurodegeneration and dementia were differentially compromised in their abilities to stabilize MT disassembly intermediates. Taken together, we propose that tau promotes the formation/stabilization of intermediate states in MT assembly and disassembly by promoting both longitudinal and lateral tubulin-tubulin contacts. We hypothesize that these activities represent fundamental aspects of tau action that normally occur at the GTP-rich ends of GTP/GDP MTs and that may be compromised in neurodegeneration-causing tau variants.
Collapse
Affiliation(s)
- Rebecca L Best
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Nichole E LaPointe
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Jiahao Liang
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Kevin Ruan
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Madeleine F Shade
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Leslie Wilson
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Stuart C Feinstein
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106.
| |
Collapse
|
26
|
Malacrida A, Meregalli C, Rodriguez-Menendez V, Nicolini G. Chemotherapy-Induced Peripheral Neuropathy and Changes in Cytoskeleton. Int J Mol Sci 2019; 20:E2287. [PMID: 31075828 PMCID: PMC6540147 DOI: 10.3390/ijms20092287] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/23/2022] Open
Abstract
Despite the different antineoplastic mechanisms of action, peripheral neurotoxicity induced by all chemotherapy drugs (anti-tubulin agents, platinum compounds, proteasome inhibitors, thalidomide) is associated with neuron morphological changes ascribable to cytoskeleton modifications. The "dying back" degeneration of distal terminals (sensory nerves) of dorsal root ganglia sensory neurons, observed in animal models, in in vitro cultures and biopsies of patients is the most evident hallmark of the perturbation of the cytoskeleton. On the other hand, in highly polarized cells like neurons, the cytoskeleton carries out its role not only in axons but also has a fundamental role in dendrite plasticity and in the organization of soma. In the literature, there are many studies focused on the antineoplastic-induced alteration of microtubule organization (and consequently, fast axonal transport defects) while very few studies have investigated the effect of the different classes of drugs on microfilaments, intermediate filaments and associated proteins. Therefore, in this review, we will focus on: (1) Highlighting the fundamental role of the crosstalk among the three filamentous subsystems and (2) investigating pivotal cytoskeleton-associated proteins.
Collapse
Affiliation(s)
- Alessio Malacrida
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| | - Cristina Meregalli
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| | - Virginia Rodriguez-Menendez
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| | - Gabriella Nicolini
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| |
Collapse
|
27
|
Castellani RJ, Perry G. Tau Biology, Tauopathy, Traumatic Brain Injury, and Diagnostic Challenges. J Alzheimers Dis 2019; 67:447-467. [PMID: 30584140 PMCID: PMC6398540 DOI: 10.3233/jad-180721] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
There is considerable interest in the pathobiology of tau protein, given its potential role in neurodegenerative diseases and aging. Tau is an important microtubule associated protein, required for the assembly of tubulin into microtubules and maintaining structural integrity of axons. Tau has other diverse cellular functions involving signal transduction, cellular proliferation, developmental neurobiology, neuroplasticity, and synaptic activity. Alternative splicing results in tau isoforms with differing microtubule binding affinity, differing representation in pathological inclusions in certain disease states, and differing roles in developmental biology and homeostasis. Tau haplotypes confer differing susceptibility to neurodegeneration. Tau phosphorylation is a normal metabolic process, critical in controlling tau's binding to microtubules, and is ongoing within the brain at all times. Tau may be hyperphosphorylated, and may aggregate as detectable fibrillar deposits in tissues, in both aging and neurodegenerative disease. The hypothesis that p-tau is neurotoxic has prompted constructs related to isomers, low-n assembly intermediates or oligomers, and the "tau prion". Human postmortem studies have elucidated broad patterns of tauopathy, with tendencies for those patterns to differ as a function of disease phenotype. However, there is extensive overlap, not only between genuine neurodegenerative diseases, but also between aging and disease. Recent studies highlight uniqueness to pathological patterns, including a pattern attributed to repetitive head trauma, although clinical correlations have been elusive. The diagnostic process for tauopathies and neurodegenerative diseases in general is challenging in many respects, and may be particularly problematic for postmortem evaluation of former athletes and military service members.
Collapse
Affiliation(s)
- Rudy J. Castellani
- Departments of Pathology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - George Perry
- College of Sciences, University of Texas, San Antonio, San Antonio, TX, USA
| |
Collapse
|
28
|
Kuznetsov IA, Kuznetsov AV. Investigating sensitivity coefficients characterizing the response of a model of tau protein transport in an axon to model parameters. Comput Methods Biomech Biomed Engin 2018; 22:71-83. [PMID: 30580604 DOI: 10.1080/10255842.2018.1534233] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Evaluating the sensitivity of biological models to various model parameters is a critical step towards advancing our understanding of biological systems. In this paper, we investigated sensitivity coefficients for a model simulating transport of tau protein along the axon. This is an important problem due to the relevance of tau transport and agglomeration to Alzheimer's disease and other tauopathies, such as some forms of parkinsonism. The sensitivity coefficients that we obtained characterize how strongly three observables (the tau concentration, average tau velocity, and the percentage of tau bound to microtubules) depend on model parameters. The fact that the observables strongly depend on a parameter characterizing tau transition from the retrograde to the anterograde kinetic states suggests the importance of motor-driven transport of tau. The observables are sensitive to kinetic constants characterizing tau concentration in the free (cytosolic) state only at small distances from the soma. Cytosolic tau can only be transported by diffusion, suggesting that diffusion-driven transport of tau only plays a role in the proximal axon. Our analysis also shows the location in the axon in which an observable has the greatest sensitivity to a certain parameter. For most parameters, this location is in the proximal axon. This could be useful for designing an experiment aimed at determining the value of this parameter. We also analyzed sensitivity of the average tau velocity, the total tau concentration, and the percentage of microtubule-bound tau to cytosolic diffusivity of tau and diffusivity of bound tau along the MT lattice. The model predicts that at small distances from the soma the effect of these two diffusion processes is comparable.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- a Perelman School of Medicine , University of Pennsylvania , Philadelphia , PA , USA.,b Department of Bioengineering , University of Pennsylvania , Philadelphia , PA , USA
| | - Andrey V Kuznetsov
- c Department of Mechanical and Aerospace Engineering , North Carolina State University , Raleigh , NC , USA
| |
Collapse
|
29
|
Kuznetsov IA, Kuznetsov AV. Simulating the effect of formation of amyloid plaques on aggregation of tau protein. Proc Math Phys Eng Sci 2018; 474:20180511. [PMID: 30602936 PMCID: PMC6304026 DOI: 10.1098/rspa.2018.0511] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022] Open
Abstract
In this paper, we develop a mathematical model that enables the investigation of the production and intracellular transport of amyloid precursor protein (APP) and tau protein in a neuron. We also investigate the aggregation of APP fragments into amyloid-β (Aβ) as well as tau aggregation into tau oligomers and neurofibrillary tangles. Using the developed model, we investigate how Aβ aggregation can influence tau transport and aggregation in both the soma and the axon. We couple the Aβ and tau agglomeration processes by assuming that the value of the kinetic constant that describes the autocatalytic growth (self-replication) reaction step of tau aggregation is proportional to the Aβ concentration. The model predicts that APP and tau are distributed differently in the axon. While APP has a uniform distribution along the axon, tau's concentration first decreases and then increases towards the synapse. Aβ is uniformly produced along the axon while misfolded tau protein is mostly produced in the proximal axon. The number of Aβ and tau polymers originating from the axon is much smaller than the number of Aβ and tau polymers originating from the soma. The rate of production of misfolded tau polymers depends on how strongly their production is facilitated by Aβ.
Collapse
Affiliation(s)
- I. A. Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A. V. Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA
| |
Collapse
|
30
|
Pearce SP, Heil M, Jensen OE, Jones GW, Prokop A. Curvature-Sensitive Kinesin Binding Can Explain Microtubule Ring Formation and Reveals Chaotic Dynamics in a Mathematical Model. Bull Math Biol 2018; 80:3002-3022. [DOI: 10.1007/s11538-018-0505-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/01/2018] [Indexed: 11/24/2022]
|
31
|
Forer A, Sheykhani R, Berns MW. Anaphase Chromosomes in Crane-Fly Spermatocytes Treated With Taxol (Paclitaxel) Accelerate When Their Kinetochore Microtubules Are Cut: Evidence for Spindle Matrix Involvement With Spindle Forces. Front Cell Dev Biol 2018; 6:77. [PMID: 30087895 PMCID: PMC6066604 DOI: 10.3389/fcell.2018.00077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/29/2018] [Indexed: 01/01/2023] Open
Abstract
Various experiments have indicated that anaphase chromosomes continue to move after their kinetochore microtubules are severed. The chromosomes move poleward at an accelerated rate after the microtubules are cut but they slow down 1-3 min later and move poleward at near the original speed. There are two published interpretations of chromosome movements with severed kinetochore microtubules. One interpretation is that dynein relocates to the severed microtubule ends and propels them poleward by pushing against non-kinetochore microtubules. The other interpretation is that components of a putative "spindle matrix" normally push kinetochore microtubules poleward and continue to do so after the microtubules are severed from the pole. In this study we distinguish between these interpretations by treating cells with taxol. Taxol eliminates microtubule dynamics, alters spindle microtubule arrangements, and inhibits dynein motor activity in vivo. If the dynein interpretation is correct, taxol should interfere with chromosome movements after kinetochore microtubules are severed because it alters the arrangements of spindle microtubules and because it blocks dynein activity. If the "spindle matrix" interpretation is correct, on the other hand, taxol should not interfere with the accelerated movements. Our results support the spindle matrix interpretation: anaphase chromosomes in taxol-treated crane-fly spermatocytes accelerated after their kinetochore microtubules were severed.
Collapse
Affiliation(s)
- Arthur Forer
- Biology Department, York University, North York, ON, Canada
| | | | - Michael W Berns
- Beckman Laser Institute and Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States.,Department of Bioengineering and Institute for Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
32
|
Monroy BY, Sawyer DL, Ackermann BE, Borden MM, Tan TC, Ori-McKenney KM. Competition between microtubule-associated proteins directs motor transport. Nat Commun 2018; 9:1487. [PMID: 29662074 PMCID: PMC5902456 DOI: 10.1038/s41467-018-03909-2] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 03/21/2018] [Indexed: 11/09/2022] Open
Abstract
Within cells, motor and non-motor microtubule-associated proteins (MAPs) simultaneously converge on the microtubule. How the binding activities of non-motor MAPs are coordinated and how they contribute to the balance and distribution of motor transport is unknown. Here, we examine the relationship between MAP7 and tau owing to their antagonistic roles in vivo. We find that MAP7 and tau compete for binding to microtubules, and determine a mechanism by which MAP7 displaces tau from the lattice. MAP7 promotes kinesin-based transport in vivo and strongly recruits kinesin-1 to the microtubule in vitro, providing evidence for direct enhancement of motor motility by a MAP. Both MAP7 and tau strongly inhibit kinesin-3 and have no effect on cytoplasmic dynein, demonstrating that MAPs differentially control distinct classes of motors. Overall, these results reveal a general principle for how MAP competition dictates access to the microtubule to determine the correct distribution and balance of motor activity.
Collapse
Affiliation(s)
- Brigette Y Monroy
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Danielle L Sawyer
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Bryce E Ackermann
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Melissa M Borden
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Tracy C Tan
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Kassandra M Ori-McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
33
|
Kuznetsov IA, Kuznetsov AV. How the formation of amyloid plaques and neurofibrillary tangles may be related: a mathematical modelling study. Proc Math Phys Eng Sci 2018; 474:20170777. [PMID: 29507520 PMCID: PMC5832841 DOI: 10.1098/rspa.2017.0777] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/12/2018] [Indexed: 12/12/2022] Open
Abstract
We develop a mathematical model that enables us to investigate possible mechanisms by which two primary markers of Alzheimer's disease (AD), extracellular amyloid plaques and intracellular tangles, may be related. Our model investigates the possibility that the decay of anterograde axonal transport of amyloid precursor protein (APP), caused by toxic tau aggregates, leads to decreased APP transport towards the synapse and APP accumulation in the soma. The developed model thus couples three processes: (i) slow axonal transport of tau, (ii) tau misfolding and agglomeration, which we simulated by using the Finke-Watzky model and (iii) fast axonal transport of APP. Because the timescale for tau agglomeration is much larger than that for tau transport, we suggest using the quasi-steady-state approximation for formulating and solving the governing equations for these three processes. Our results suggest that misfolded tau most likely accumulates in the beginning of the axon. The analysis of APP transport suggests that APP will also likely accumulate in the beginning of the axon, causing an increased APP concentration in this region, which could be interpreted as a 'traffic jam'. The APP flux towards the synapse is significantly reduced by tau misfolding, but not due to the APP traffic jam, which can be viewed as a symptom, but rather due to the reduced affinity of kinesin-1 motors to APP-transporting vesicles.
Collapse
Affiliation(s)
- I. A. Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A. V. Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695–7910, USA
| |
Collapse
|
34
|
Balabanian L, Berger CL, Hendricks AG. Acetylated Microtubules Are Preferentially Bundled Leading to Enhanced Kinesin-1 Motility. Biophys J 2017; 113:1551-1560. [PMID: 28978447 DOI: 10.1016/j.bpj.2017.08.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022] Open
Abstract
The motor proteins kinesin and dynein transport organelles, mRNA, proteins, and signaling molecules along the microtubule cytoskeleton. In addition to serving as tracks for transport, the microtubule cytoskeleton directs intracellular trafficking by regulating the activity of motor proteins through the organization of the filament network, microtubule-associated proteins, and tubulin posttranslational modifications. However, it is not well understood how these factors influence motor motility, and in vitro assays and live cell observations often produce disparate results. To systematically examine the factors that contribute to cytoskeleton-based regulation of motor protein motility, we extracted intact microtubule networks from cells and tracked the motility of single fluorescently labeled motor proteins on these cytoskeletons. We find that tubulin acetylation alone does not directly affect kinesin-1 motility. However, acetylated microtubules are predominantly bundled, and bundling enhances kinesin run lengths and provides a greater number of available kinesin binding sites. The neuronal MAP tau is also not sensitive to tubulin acetylation, but enriches preferentially on highly curved regions of microtubules where it strongly inhibits kinesin motility. Taken together, these results suggest that the organization of the microtubule network is a key contributor to the regulation of motor-based transport.
Collapse
Affiliation(s)
- Linda Balabanian
- Department of Bioengineering, McGill University, Montreal, Québec, Canada
| | - Christopher L Berger
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| | - Adam G Hendricks
- Department of Bioengineering, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
35
|
Ramkumar A, Jong BY, Ori-McKenney KM. ReMAPping the microtubule landscape: How phosphorylation dictates the activities of microtubule-associated proteins. Dev Dyn 2017; 247:138-155. [PMID: 28980356 DOI: 10.1002/dvdy.24599] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Classical microtubule-associated proteins (MAPs) were originally identified based on their co-purification with microtubules assembled from mammalian brain lysate. They have since been found to perform a range of functions involved in regulating the dynamics of the microtubule cytoskeleton. Most of these MAPs play integral roles in microtubule organization during neuronal development, microtubule remodeling during neuronal activity, and microtubule stabilization during neuronal maintenance. As a result, mutations in MAPs contribute to neurodevelopmental disorders, psychiatric conditions, and neurodegenerative diseases. MAPs are post-translationally regulated by phosphorylation depending on developmental time point and cellular context. Phosphorylation can affect the microtubule affinity, cellular localization, or overall function of a particular MAP and can thus have profound implications for neuronal health. Here we review MAP1, MAP2, MAP4, MAP6, MAP7, MAP9, tau, and DCX, and how each is regulated by phosphorylation in neuronal physiology and disease. Developmental Dynamics 247:138-155, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amrita Ramkumar
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | - Brigette Y Jong
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | | |
Collapse
|
36
|
Kneynsberg A, Combs B, Christensen K, Morfini G, Kanaan NM. Axonal Degeneration in Tauopathies: Disease Relevance and Underlying Mechanisms. Front Neurosci 2017; 11:572. [PMID: 29089864 PMCID: PMC5651019 DOI: 10.3389/fnins.2017.00572] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/29/2017] [Indexed: 12/14/2022] Open
Abstract
Tauopathies are a diverse group of diseases featuring progressive dying-back neurodegeneration of specific neuronal populations in association with accumulation of abnormal forms of the microtubule-associated protein tau. It is well-established that the clinical symptoms characteristic of tauopathies correlate with deficits in synaptic function and neuritic connectivity early in the course of disease, but mechanisms underlying these critical pathogenic events are not fully understood. Biochemical in vitro evidence fueled the widespread notion that microtubule stabilization represents tau's primary biological role and that the marked atrophy of neurites observed in tauopathies results from loss of microtubule stability. However, this notion contrasts with the mild phenotype associated with tau deletion. Instead, an analysis of cellular hallmarks common to different tauopathies, including aberrant patterns of protein phosphorylation and early degeneration of axons, suggests that alterations in kinase-based signaling pathways and deficits in axonal transport (AT) associated with such alterations contribute to the loss of neuronal connectivity triggered by pathogenic forms of tau. Here, we review a body of literature providing evidence that axonal pathology represents an early and common pathogenic event among human tauopathies. Observations of axonal degeneration in animal models of specific tauopathies are discussed and similarities to human disease highlighted. Finally, we discuss potential mechanistic pathways other than microtubule destabilization by which disease-related forms of tau may promote axonopathy.
Collapse
Affiliation(s)
- Andrew Kneynsberg
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Benjamin Combs
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Kyle Christensen
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Nicholas M Kanaan
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|
37
|
Kuznetsov IA, Kuznetsov AV. What mechanisms of tau protein transport could be responsible for the inverted tau concentration gradient in degenerating axons? MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2017; 34:125-150. [PMID: 27034421 DOI: 10.1093/imammb/dqv041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 11/30/2015] [Indexed: 12/27/2022]
Abstract
In tauopathies, such as Alzheimer's disease (AD), microtubule (MT)-associated protein tau detaches from MTs and aggregates, eventually forming insoluble neurofibrillary tangles. In a healthy axon, the tau concentration increases toward the axon terminal, but in a degenerating axon, the tau concentration gradient is inverted and the highest tau concentration is in the soma. In this article, we developed a mathematical model of tau transport in axons. We calibrated and tested the model by using published distributions of tau concentration and tau average velocity in a healthy axon. According to published research, the inverted tau concentration gradient may be one of the reasons leading to AD. We therefore used the model to investigate what modifications in tau transport can lead to the inverted tau concentration gradient. We investigated whether tau detachment from MTs due to tau hyperphosphorylation can cause the inverted tau concentration gradient. We found that the assumption that most tau molecules are detached from MTs does not consistently predict the inverted tau concentration gradient; the predicted tau distribution becomes more uniform if the axon length is increased. We then hypothesized that in degenerating axons some tau remains bound to MTs and participates in the component 'a' of slow axonal transport but that the rate of tau reversals from anterograde to retrograde motion increases. We demonstrated that this hypothesis results in a tau distribution where the tau concentration has its maximum value at the axon hillock and its minimum value at the axon terminal, in agreement with what is observed in AD. Our results thus suggest that defects in active transport of tau may be a contributing factor to the onset of neural degeneration.
Collapse
|
38
|
Dynamic microtubule association of Doublecortin X (DCX) is regulated by its C-terminus. Sci Rep 2017; 7:5245. [PMID: 28701724 PMCID: PMC5507856 DOI: 10.1038/s41598-017-05340-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022] Open
Abstract
Doublecortin X (DCX), known to be essential for neuronal migration and cortical layering in the developing brain, is a 40 kDa microtubule (MT)-associated protein. DCX directly interacts with MTs via its two structured doublecortin (DC) domains, but the dynamics of this association and the possible regulatory roles played by the flanking unstructured regions remain poorly defined. Here, we employ quantitative fluorescence recovery after photobleaching (FRAP) protocols in living cells to reveal that DCX shows remarkably rapid and complete exchange within the MT network but that the removal of the C-terminal region significantly slows this exchange. We further probed how MT organization or external stimuli could additionally modulate DCX exchange dynamics. MT depolymerisation (nocodazole treatment) or stabilization (taxol treatment) further enhanced DCX exchange rates, however the exchange rates for the C-terminal truncated DCX protein were resistant to the impact of taxol-induced stabilization. Furthermore, in response to a hyperosmotic stress stimulus, DCX exchange dynamics were slowed, and again the C-terminal truncated DCX protein was resistant to the stimulus. Thus, the DCX dynamically associates with MTs in living cells and its C-terminal region plays important roles in the MT-DCX association.
Collapse
|
39
|
Kuznetsov IA, Kuznetsov AV. Simulating tubulin-associated unit transport in an axon: using bootstrapping for estimating confidence intervals of best-fit parameter values obtained from indirect experimental data. Proc Math Phys Eng Sci 2017; 473:20170045. [PMID: 28588409 DOI: 10.1098/rspa.2017.0045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/03/2017] [Indexed: 02/06/2023] Open
Abstract
In this paper, we first develop a model of axonal transport of tubulin-associated unit (tau) protein. We determine the minimum number of parameters necessary to reproduce published experimental results, reducing the number of parameters from 18 in the full model to eight in the simplified model. We then address the following questions: Is it possible to estimate parameter values for this model using the very limited amount of published experimental data? Furthermore, is it possible to estimate confidence intervals for the determined parameters? The idea that is explored in this paper is based on using bootstrapping. Model parameters were estimated by minimizing the objective function that simulates the discrepancy between the model predictions and experimental data. Residuals were then identified by calculating the differences between the experimental data and model predictions. New, surrogate 'experimental' data were generated by randomly resampling residuals. By finding sets of best-fit parameters for a large number of surrogate data the histograms for the model parameters were produced. These histograms were then used to estimate confidence intervals for the model parameters, by using the percentile bootstrap. Once the model was calibrated, we applied it to analysing some features of tau transport that are not accessible to current experimental techniques.
Collapse
Affiliation(s)
- I A Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA
| |
Collapse
|
40
|
Duan AR, Jonasson EM, Alberico EO, Li C, Scripture JP, Miller RA, Alber MS, Goodson HV. Interactions between Tau and Different Conformations of Tubulin: Implications for Tau Function and Mechanism. J Mol Biol 2017; 429:1424-1438. [PMID: 28322917 DOI: 10.1016/j.jmb.2017.03.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/22/2017] [Accepted: 03/12/2017] [Indexed: 11/16/2022]
Abstract
Tau is a multifaceted neuronal protein that stabilizes microtubules (MTs), but the mechanism of this activity remains poorly understood. Questions include whether Tau binds MTs laterally or longitudinally and whether Tau's binding affinity depends on the nucleotide state of tubulin. We observed that Tau binds tightly to Dolastatin-10 tubulin rings and promotes the formation of Dolastatin-10 ring stacks, implying that Tau can crosslink MT protofilaments laterally. In addition, we found that Tau prefers GDP-like tubulin conformations, which implies that Tau binding to the MT surface is biased away from the dynamic GTP-rich MT tip. To investigate the potential impact of these Tau activities on MT stabilization, we incorporated them into our previously developed dimer-scale computational model of MT dynamics. We found that lateral crosslinking activities have a much greater effect on MT stability than do longitudinal crosslinking activities, and that introducing a bias toward GDP tubulin has little impact on the observed MT stabilization. To address the question of why Tau is GDP-tubulin-biased, we tested whether Tau might affect MT binding of the +TIP EB1. We confirmed recent reports that Tau binds directly to EB1 and that Tau competes with EB1 for MT binding. Our results lead to a conceptual model where Tau stabilizes the MT lattice by strengthening lateral interactions between protofilaments. We propose that Tau's GDP preference allows the cell to independently regulate the dynamics of the MT tip and the stability of the lattice.
Collapse
Affiliation(s)
- Aranda R Duan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Erin M Jonasson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Emily O Alberico
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Chunlei Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jared P Scripture
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel A Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Mark S Alber
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Mathematics, University of California, Riverside, CA 92521, USA
| | - Holly V Goodson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
41
|
Bakota L, Ussif A, Jeserich G, Brandt R. Systemic and network functions of the microtubule-associated protein tau: Implications for tau-based therapies. Mol Cell Neurosci 2017; 84:132-141. [PMID: 28318914 DOI: 10.1016/j.mcn.2017.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/23/2017] [Accepted: 03/05/2017] [Indexed: 01/04/2023] Open
Abstract
Tau is a microtubule-associated neuronal protein, whose primary role was long thought to regulate axonal microtubule assembly. Tau is subject to many posttranslational modifications and can aggregate into neurofibrillary tangles, which are considered to be a hallmark of several neurodegenerative diseases collectively called "tauopathies". The most common tauopathy is Alzheimer's disease, where tau pathology correlates with sites of neurodegeneration. Tau belongs to the class of intrinsically disordered proteins, which are known to interact with many partners and are considered to be involved in various signaling, regulation and recognition processes. Thus more recent evidence indicates that tau functionally interacts with many proteins and different cellular structures, which may have an important physiological role and may be involved in neurodegenerative processes. Furthermore, tau can be released from neurons and exert functional effects on other cells. This review article weighs the evidence that tau has subtle but important systemic effects on neuronal network function by maintaining physiological neuronal transmission and synaptic plasticity, which are possibly independent from tau's microtubule modulating activities. Implications for tau-based therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Abdala Ussif
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Gunnar Jeserich
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany.
| |
Collapse
|
42
|
Back to the tubule: microtubule dynamics in Parkinson's disease. Cell Mol Life Sci 2016; 74:409-434. [PMID: 27600680 PMCID: PMC5241350 DOI: 10.1007/s00018-016-2351-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
Cytoskeletal homeostasis is essential for the development, survival and maintenance of an efficient nervous system. Microtubules are highly dynamic polymers important for neuronal growth, morphology, migration and polarity. In cooperation with several classes of binding proteins, microtubules regulate long-distance intracellular cargo trafficking along axons and dendrites. The importance of a delicate interplay between cytoskeletal components is reflected in several human neurodegenerative disorders linked to abnormal microtubule dynamics, including Parkinson’s disease (PD). Mounting evidence now suggests PD pathogenesis might be underlined by early cytoskeletal dysfunction. Advances in genetics have identified PD-associated mutations and variants in genes encoding various proteins affecting microtubule function including the microtubule-associated protein tau. In this review, we highlight the role of microtubules, their major posttranslational modifications and microtubule associated proteins in neuronal function. We then present key evidence on the contribution of microtubule dysfunction to PD. Finally, we discuss how regulation of microtubule dynamics with microtubule-targeting agents and deacetylase inhibitors represents a promising strategy for innovative therapeutic development.
Collapse
|
43
|
Arendt T, Stieler JT, Holzer M. Tau and tauopathies. Brain Res Bull 2016; 126:238-292. [PMID: 27615390 DOI: 10.1016/j.brainresbull.2016.08.018] [Citation(s) in RCA: 424] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022]
|
44
|
Niewidok B, Igaev M, Sündermann F, Janning D, Bakota L, Brandt R. Presence of a carboxy-terminal pseudorepeat and disease-like pseudohyperphosphorylation critically influence tau's interaction with microtubules in axon-like processes. Mol Biol Cell 2016; 27:3537-3549. [PMID: 27582388 PMCID: PMC5221586 DOI: 10.1091/mbc.e16-06-0402] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/24/2016] [Indexed: 12/31/2022] Open
Abstract
A refined FDAP approach is used to analyze tau’s behavior in axon-like processes. A conserved C-terminal pseudorepeat and disease-like pseudohyperphosphorylation critically influence tau’s microtubule interaction. The results contribute to an understanding of pathological processes that lead to tau’s redistribution during disease. A current challenge of cell biology is to investigate molecular interactions in subcellular compartments of living cells to overcome the artificial character of in vitro studies. To dissect the interaction of the neuronal microtubule (MT)-associated protein tau with MTs in axon-like processes, we used a refined fluorescence decay after photoactivation approach and single-molecule tracking. We found that isoform variation had only a minor influence on the tau–MT interaction, whereas the presence of a C-terminal pseudorepeat region (PRR) greatly increased MT binding by a greater-than-sixfold reduction of the dissociation rate. Bioinformatic analysis revealed that the PRR contained a highly conserved motif of 18 amino acids. Disease-associated tau mutations in the PRR (K369I, G389R) did not influence apparent MT binding but increased its dynamicity. Simulation of disease-like tau hyperphosphorylation dramatically diminished the tau–MT interaction by a greater-than-fivefold decrease of the association rate with no major change in the dissociation rate. Apparent binding of tau to MTs was similar in axons and dendrites but more sensitive to increased phosphorylation in axons. Our data indicate that under the conditions of high MT density that prevail in the axon, tau’s MT binding and localization are crucially affected by the presence of the PRR and tau hyperphosphorylation.
Collapse
Affiliation(s)
- Benedikt Niewidok
- Department of Neurobiology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Maxim Igaev
- Department of Neurobiology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Frederik Sündermann
- Department of Neurobiology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Dennis Janning
- Department of Neurobiology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, 49076 Osnabrück, Germany
| |
Collapse
|
45
|
Sohn PD, Tracy TE, Son HI, Zhou Y, Leite REP, Miller BL, Seeley WW, Grinberg LT, Gan L. Acetylated tau destabilizes the cytoskeleton in the axon initial segment and is mislocalized to the somatodendritic compartment. Mol Neurodegener 2016; 11:47. [PMID: 27356871 PMCID: PMC4928318 DOI: 10.1186/s13024-016-0109-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 06/01/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Neurons are highly polarized cells in which asymmetric axonal-dendritic distribution of proteins is crucial for neuronal function. Loss of polarized distribution of the axonal protein tau is an early sign of Alzheimer's disease (AD) and other neurodegenerative disorders. The cytoskeletal network in the axon initial segment (AIS) forms a barrier between the axon and the somatodentritic compartment, contributing to axonal retention of tau. Although perturbation of the AIS cytoskeleton has been implicated in neurological disorders, the molecular triggers and functional consequence of AIS perturbation are incompletely understood. RESULTS Here we report that tau acetylation and consequent destabilization of the AIS cytoskeleton promote the somatodendritic mislocalization of tau. AIS cytoskeletal proteins, including ankyrin G and βIV-spectrin, were downregulated in AD brains and negatively correlated with an increase in tau acetylated at K274 and K281. AIS proteins were also diminished in transgenic mice expressing tauK274/281Q, a tau mutant that mimics K274 and K281 acetylation. In primary neuronal cultures, the tauK274/281Q mutant caused hyperdynamic microtubules (MTs) in the AIS, shown by live-imaging of MT mobility and fluorescence recovery after photobleaching. Using photoconvertible tau constructs, we found that axonal tauK274/281Q was missorted into the somatodendritic compartment. Stabilizing MTs with epothilone D to restore the cytoskeletal barrier in the AIS prevented tau mislocalization in primary neuronal cultures. CONCLUSIONS Together, these findings demonstrate that tau acetylation contributes to the pathogenesis of neurodegenerative disease by compromising the cytoskeletal sorting machinery in the AIS.
Collapse
Affiliation(s)
- Peter Dongmin Sohn
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, 94158, USA
| | - Tara E Tracy
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Hye-In Son
- Gladstone Institute of Virology & Immunology, San Francisco, CA, 94158, USA
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA
| | - Renata E P Leite
- Physiopathology in Aging Lab/Brazilian Aging Brain Study Group-LIM22, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology University of California, San Francisco, CA, 94158, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology University of California, San Francisco, CA, 94158, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology University of California, San Francisco, CA, 94158, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA.
- Neuroscience Graduate Program, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
46
|
Doublecortin Is Excluded from Growing Microtubule Ends and Recognizes the GDP-Microtubule Lattice. Curr Biol 2016; 26:1549-1555. [PMID: 27238282 DOI: 10.1016/j.cub.2016.04.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 03/03/2016] [Accepted: 04/08/2016] [Indexed: 12/14/2022]
Abstract
Many microtubule (MT) functions are mediated by a diverse class of proteins (+TIPs) at growing MT plus ends that control intracellular MT interactions and dynamics and depend on end-binding proteins (EBs) [1]. Cryoelectron microscopy has recently identified the EB binding site as the interface of four tubulin dimers that undergoes a conformational change in response to β-tubulin GTP hydrolysis [2, 3]. Doublecortin (DCX), a MT-associated protein (MAP) required for neuronal migration during cortical development [4, 5], binds to the same site as EBs [6], and recent in vitro studies proposed DCX localization to growing MT ends independent of EBs [7]. Because this conflicts with observations in neurons [8, 9] and the molecular function of DCX is not well understood, we revisited intracellular DCX dynamics at low expression levels. Here, we report that DCX is not a +TIP in cells but, on the contrary, is excluded from the EB1 domain. In addition, we find that DCX-MT interactions are highly sensitive to MT geometry. In cells, DCX binding was greatly reduced at MT segments with high local curvature. Remarkably, this geometry-dependent binding to MTs was completely reversed in the presence of taxanes, which reconciles incompatible observations in cells [9] and in vitro [10]. We propose a model explaining DCX specificity for different MT geometries based on structural changes induced by GTP hydrolysis that decreases the spacing between adjacent tubulin dimers [11]. Our data are consistent with a unique mode of MT interaction in which DCX specifically recognizes this compacted GDP-like MT lattice.
Collapse
|
47
|
Torres-Cruz FM, Rodríguez-Cruz F, Escobar-Herrera J, Barragán-Andrade N, Basurto-Islas G, Ripova D, Ávila J, Garcia-Sierra F. Expression of Tau Produces Aberrant Plasma Membrane Blebbing in Glial Cells Through RhoA-ROCK-Dependent F-Actin Remodeling. J Alzheimers Dis 2016; 52:463-82. [DOI: 10.3233/jad-150396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Francisco M. Torres-Cruz
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | - Fanny Rodríguez-Cruz
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | - Jaime Escobar-Herrera
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | - Norma Barragán-Andrade
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | | | - Daniela Ripova
- National Institute of Mental Health, Klecany, Czech Republic
| | - Jesús Ávila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM) Universidad Autónoma de Madrid, Spain
| | - Francisco Garcia-Sierra
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
48
|
Liebert AD, Chow RT, Bicknell BT, Varigos E. Neuroprotective Effects Against POCD by Photobiomodulation: Evidence from Assembly/Disassembly of the Cytoskeleton. J Exp Neurosci 2016; 10:1-19. [PMID: 26848276 PMCID: PMC4737522 DOI: 10.4137/jen.s33444] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/09/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a decline in memory following anaesthesia and surgery in elderly patients. While often reversible, it consumes medical resources, compromises patient well-being, and possibly accelerates progression into Alzheimer's disease. Anesthetics have been implicated in POCD, as has neuroinflammation, as indicated by cytokine inflammatory markers. Photobiomodulation (PBM) is an effective treatment for a number of conditions, including inflammation. PBM also has a direct effect on microtubule disassembly in neurons with the formation of small, reversible varicosities, which cause neural blockade and alleviation of pain symptoms. This mimics endogenously formed varicosities that are neuroprotective against damage, toxins, and the formation of larger, destructive varicosities and focal swellings. It is proposed that PBM may be effective as a preconditioning treatment against POCD; similar to the PBM treatment, protective and abscopal effects that have been demonstrated in experimental models of macular degeneration, neurological, and cardiac conditions.
Collapse
Affiliation(s)
| | - Roberta T. Chow
- Brain and Mind Institute, University of Sydney, Sydney, NSW, Australia
| | | | | |
Collapse
|
49
|
Penazzi L, Tackenberg C, Ghori A, Golovyashkina N, Niewidok B, Selle K, Ballatore C, Smith AB, Bakota L, Brandt R. Aβ-mediated spine changes in the hippocampus are microtubule-dependent and can be reversed by a subnanomolar concentration of the microtubule-stabilizing agent epothilone D. Neuropharmacology 2016; 105:84-95. [PMID: 26772969 DOI: 10.1016/j.neuropharm.2016.01.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/10/2015] [Accepted: 01/03/2016] [Indexed: 10/25/2022]
Abstract
Dendritic spines represent the major postsynaptic input of excitatory synapses. Loss of spines and changes in their morphology correlate with cognitive impairment in Alzheimer's disease (AD) and are thought to occur early during pathology. Therapeutic intervention at a preclinical stage of AD to modify spine changes might thus be warranted. To follow the development and to potentially interfere with spine changes over time, we established a long term ex vivo model from organotypic cultures of the hippocampus from APP transgenic and control mice. The cultures exhibit spine loss in principal hippocampal neurons, which closely resembles the changes occurring in vivo, and spine morphology progressively changes from mushroom-shaped to stubby. We demonstrate that spine changes are completely reversed within few days after blocking amyloid-β (Aβ) production with the gamma-secretase inhibitor DAPT. We show that the microtubule disrupting drug nocodazole leads to spine loss similar to Aβ expressing cultures and suppresses DAPT-mediated spine recovery in slices from APP transgenic mice. Finally, we report that epothilone D (EpoD) at a subnanomolar concentration, which slightly stabilizes microtubules in model neurons, completely reverses Aβ-induced spine loss and increases thin spine density. Taken together the data indicate that Aβ causes spine changes by microtubule destabilization and that spine recovery requires microtubule polymerization. Moreover, our results suggest that a low, subtoxic concentration of EpoD is sufficient to reduce spine loss during the preclinical stage of AD.
Collapse
Affiliation(s)
- Lorène Penazzi
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Christian Tackenberg
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Adnan Ghori
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Nataliya Golovyashkina
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Benedikt Niewidok
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Karolin Selle
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Carlo Ballatore
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, United States; Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Amos B Smith
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany.
| |
Collapse
|
50
|
Axonal Transport Impairment in Chemotherapy-Induced Peripheral Neuropathy. TOXICS 2015; 3:322-341. [PMID: 29051467 PMCID: PMC5606679 DOI: 10.3390/toxics3030322] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/28/2015] [Accepted: 08/03/2015] [Indexed: 12/18/2022]
Abstract
Chemotherapy-Induced Peripheral Neuropathy (CIPN) is a dose-limiting side effect of several antineoplastic drugs which significantly reduces patients’ quality of life. Although different molecular mechanisms have been investigated, CIPN pathobiology has not been clarified yet. It has largely been recognized that Dorsal Root Ganglia are the main targets of chemotherapy and that the longest nerves are the most damaged, together with fast axonal transport. Indeed, this bidirectional cargo-specific transport has a pivotal role in neuronal function and its impairment is involved in several neurodegenerative and neurodevelopmental diseases. Literature data demonstrate that, despite different mechanisms of action, all antineoplastic agents impair the axonal trafficking to some extent and the severity of the neuropathy correlates with the degree of damage on this bidirectional transport. In this paper, we will examine the effect of the main old and new chemotherapeutic drug categories on axonal transport, with the aim of clarifying their potential mechanisms of action, and, if possible, of identifying neuroprotective strategies, based on the knowledge of the alterations induced by each drugs.
Collapse
|