1
|
Asada-Utsugi M, Urushitani M. Tau beyond Tangles: DNA Damage Response and Cytoskeletal Protein Crosstalk on Neurodegeneration. Int J Mol Sci 2024; 25:7906. [PMID: 39063148 PMCID: PMC11277103 DOI: 10.3390/ijms25147906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Neurons in the brain are continuously exposed to various sources of DNA damage. Although the mechanisms of DNA damage repair in mitotic cells have been extensively characterized, the repair pathways in post-mitotic neurons are still largely elusive. Moreover, inaccurate repair can result in deleterious mutations, including deletions, insertions, and chromosomal translocations, ultimately compromising genomic stability. Since neurons are terminally differentiated cells, they cannot employ homologous recombination (HR) for double-strand break (DSB) repair, suggesting the existence of neuron-specific repair mechanisms. Our research has centered on the microtubule-associated protein tau (MAPT), a crucial pathological protein implicated in neurodegenerative diseases, and its interplay with neurons' DNA damage response (DDR). This review aims to provide an updated synthesis of the current understanding of the complex interplay between DDR and cytoskeletal proteins in neurons, with a particular focus on the role of tau in neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Makoto Urushitani
- Department of Neurology, Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| |
Collapse
|
2
|
Yakout DW, Shroff A, Wei W, Thaker V, Allen ZD, Sajish M, Nazarko TY, Mabb AM. Tau regulates Arc stability in neuronal dendrites via a proteasome-sensitive but ubiquitin-independent pathway. J Biol Chem 2024; 300:107237. [PMID: 38552740 PMCID: PMC11061231 DOI: 10.1016/j.jbc.2024.107237] [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: 05/01/2023] [Revised: 02/23/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
Tauopathies are neurodegenerative disorders characterized by the deposition of aggregates of the microtubule-associated protein tau, a main component of neurofibrillary tangles. Alzheimer's disease (AD) is the most common type of tauopathy and dementia, with amyloid-beta pathology as an additional hallmark feature of the disease. Besides its role in stabilizing microtubules, tau is localized at postsynaptic sites and can regulate synaptic plasticity. The activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that plays a key role in synaptic plasticity, learning, and memory. Arc has been implicated in AD pathogenesis and regulates the release of amyloid-beta. We found that decreased Arc levels correlate with AD status and disease severity. Importantly, Arc protein was upregulated in the hippocampus of Tau KO mice and dendrites of Tau KO primary hippocampal neurons. Overexpression of tau decreased Arc stability in an activity-dependent manner, exclusively in neuronal dendrites, which was coupled to an increase in the expression of dendritic and somatic surface GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. The tau-dependent decrease in Arc was found to be proteasome-sensitive, yet independent of Arc ubiquitination and required the endophilin-binding domain of Arc. Importantly, these effects on Arc stability and GluA1 localization were not observed in the commonly studied tau mutant, P301L. These observations provide a potential molecular basis for synaptic dysfunction mediated through the accumulation of tau in dendrites. Our findings confirm that Arc is misregulated in AD and further show a physiological role for tau in regulating Arc stability and AMPA receptor targeting.
Collapse
Affiliation(s)
- Dina W Yakout
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Ankit Shroff
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Wei Wei
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Vishrut Thaker
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Zachary D Allen
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Mathew Sajish
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Taras Y Nazarko
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Angela M Mabb
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
3
|
Chang K, Barrett A, Pham K, Troncoso JC. Lateral geniculate body is spared of tau pathology in Pick disease. J Neuropathol Exp Neurol 2024; 83:238-244. [PMID: 38412343 PMCID: PMC10951972 DOI: 10.1093/jnen/nlae016] [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] [Indexed: 02/29/2024] Open
Abstract
The pathobiology of tau is of great importance for understanding the mechanisms of neurodegeneration in aging and age-associated disorders such as Alzheimer disease (AD) and frontotemporal dementias. It is critical to identify neuronal populations and brain regions that are vulnerable or resistant to tau pathological changes. Pick disease (PiD) is a three-repeat (3R) tauopathy that belongs to the group of frontotemporal lobar degenerations. The neuropathologic changes of PiD are characterized by globular tau-positive neuronal intracytoplasmic inclusions, called Pick bodies, in the granule cells of the dentate gyrus and frontal and temporal neocortices, and ballooned neurons, named Pick neurons, in the neocortex. In the present study, we examined 13 autopsy-confirmed cases of PiD. Using immunohistochemistry for phospho-tau (AT8) and 3R tau isoform, all PiD cases demonstrated extensive lesions involving the hippocampus and neocortex. However, the lateral geniculate body (LGB) is spared of significant tau lesions in contrast to the neighboring hippocampus and other thalamic nuclei. Only 1 PiD case (7.7%) had tau-positive neurons, and 4 cases had tau-positive neurites (31%) in the LGB. By contrast, the LGB does consistently harbor tau lesions in other tauopathies including progressive supranuclear palsy, corticobasal degeneration, and AD.
Collapse
Affiliation(s)
- Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Alexander Barrett
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Khoa Pham
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Sahara N, Higuchi M. Diagnostic and therapeutic targeting of pathological tau proteins in neurodegenerative disorders. FEBS Open Bio 2024; 14:165-180. [PMID: 37746832 PMCID: PMC10839408 DOI: 10.1002/2211-5463.13711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/22/2023] [Indexed: 09/26/2023] Open
Abstract
Tauopathies, characterized by fibrillar tau accumulation in neurons and glial cells, constitute a major neuropathological category of neurodegenerative diseases. Neurofibrillary tau lesions are strongly associated with cognitive deficits in these diseases, but the causal mechanisms underlying tau-induced neuronal dysfunction remain unresolved. Recent advances in cryo-electron microscopy examination have revealed various core structures of tau filaments from different tauopathy patients, which can be used to classify tauopathies. In vivo visualization of tau pathology is now available using several tau positron emission tomography tracers. Among these radioprobes, PM-PBB3 allows high-contrast imaging of tau deposits in the brains of patients with diverse disorders and tauopathy mouse models. Selective degradation of pathological tau species by the ubiquitin-proteasome system or autophagy machinery is a potential therapeutic strategy. Alternatively, the non-cell-autonomous clearance of pathological tau species through neuron-glia networks could be reinforced as a disease-modifying treatment. In addition, the development of neuroinflammatory biomarkers is required for understanding the contribution of immunocompetent cells in the brain to preventing neurodegeneration. This review provides an overview of the current research and development of diagnostic and therapeutic agents targeting divergent tau pathologies.
Collapse
Affiliation(s)
- Naruhiko Sahara
- Department of Functional Brain Imaging, Institute for Quantum Medical SciencesNational Institutes for Quantum Science and TechnologyChibaJapan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical SciencesNational Institutes for Quantum Science and TechnologyChibaJapan
| |
Collapse
|
5
|
Bowles KR, Pugh DA, Pedicone C, Oja L, Weitzman SA, Liu Y, Chen JL, Disney MD, Goate AM. Development of MAPT S305 mutation models exhibiting elevated 4R tau expression, resulting in altered neuronal and astrocytic function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543224. [PMID: 37333200 PMCID: PMC10274740 DOI: 10.1101/2023.06.02.543224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Due to the importance of 4R tau in the pathogenicity of primary tauopathies, it has been challenging to model these diseases in iPSC-derived neurons, which express very low levels of 4R tau. To address this problem we have developed a panel of isogenic iPSC lines carrying the MAPT splice-site mutations S305S, S305I or S305N, derived from four different donors. All three mutations significantly increased the proportion of 4R tau expression in iPSC-neurons and astrocytes, with up to 80% 4R transcripts in S305N neurons from as early as 4 weeks of differentiation. Transcriptomic and functional analyses of S305 mutant neurons revealed shared disruption in glutamate signaling and synaptic maturity, but divergent effects on mitochondrial bioenergetics. In iPSC-astrocytes, S305 mutations induced lysosomal disruption and inflammation and exacerbated internalization of exogenous tau that may be a precursor to the glial pathologies observed in many tauopathies. In conclusion, we present a novel panel of human iPSC lines that express unprecedented levels of 4R tau in neurons and astrocytes. These lines recapitulate previously characterized tauopathy-relevant phenotypes, but also highlight functional differences between the wild type 4R and mutant 4R proteins. We also highlight the functional importance of MAPT expression in astrocytes. These lines will be highly beneficial to tauopathy researchers enabling a more complete understanding of the pathogenic mechanisms underlying 4R tauopathies across different cell types.
Collapse
Affiliation(s)
- KR Bowles
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - DA Pugh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - C Pedicone
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - L Oja
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - SA Weitzman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Y Liu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - JL Chen
- Department of Chemistry, Scripps Research Institute, Jupiter, FL, United States of America
| | - MD Disney
- Department of Chemistry, Scripps Research Institute, Jupiter, FL, United States of America
| | - AM Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
6
|
Havlicek DF, Furhang R, Nikulina E, Smith-Salzberg B, Lawless S, Severin SA, Mallaboeva S, Nayab F, Seifert AC, Crary JF, Bergold PJ. A single closed head injury in male adult mice induces chronic, progressive white matter atrophy and increased phospho-tau expressing oligodendrocytes. Exp Neurol 2023; 359:114241. [PMID: 36240881 DOI: 10.1016/j.expneurol.2022.114241] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
Abstract
Traumatic brain injury (TBI) acutely damages the brain; this injury can evolve into chronic neurodegeneration. While much is known about the chronic effects arising from multiple mild TBIs, far less is known about the long-term effects of a single moderate to severe TBI. We found that a single moderate closed head injury to mice induces diffuse axonal injury within 1-day post-injury (DPI). At 14 DPI, injured animals have atrophy of ipsilesional cortex, thalamus, and corpus callosum, with bilateral atrophy of the dorsal fornix. Atrophy of the ipsilesional corpus callosum is accompanied by decreased fractional anisotropy and increased mean and radial diffusivity that remains unchanged between 14 and 180 DPI. Injured animals show an increased density of phospho-tau immunoreactive (pTau+) cells in the ipsilesional cortex and thalamus, and bilaterally in corpus callosum. Between 14 and 180 DPI, atrophy occurs in the ipsilesional ventral fornix, contralesional corpus callosum, and bilateral internal capsule. Diffusion tensor MRI parameters remain unchanged in white matter regions with delayed atrophy. Between 14 and 180 DPI, pTau+ cell density increases bilaterally in corpus callosum, but decreases in cortex and thalamus. The location of pTau+ cells within the ipsilesional corpus callosum changes between 14 and 180 DPI; density of all cells increases including pTau+ or pTau- cells. >90% of the pTau+ cells are in the oligodendrocyte lineage in both gray and white matter. Density of thioflavin-S+ cells in thalamus increases by 180 DPI. These data suggest a single closed head impact produces multiple forms of chronic neurodegeneration. Gray and white matter regions proximal to the impact site undergo early atrophy. More distal white matter regions undergo chronic, progressive white matter atrophy with an increasing density of oligodendrocytes containing pTau. These data suggest a complex chronic neurodegenerative process arising from a single moderate closed head injury.
Collapse
Affiliation(s)
- David F Havlicek
- School of Graduate Studies, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Rachel Furhang
- School of Graduate Studies, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Elena Nikulina
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Bayle Smith-Salzberg
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Siobhán Lawless
- School of Graduate Studies, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Sasha A Severin
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Sevara Mallaboeva
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Fizza Nayab
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Alan C Seifert
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - John F Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Peter J Bergold
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States of America.
| |
Collapse
|
7
|
Liquid-Liquid Phase Separation Promotes Protein Aggregation and Its Implications in Ferroptosis in Parkinson’s Disease Dementia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7165387. [PMID: 36246407 PMCID: PMC9560807 DOI: 10.1155/2022/7165387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/07/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022]
Abstract
The pathological features of PDD are represented by dopaminergic neuronal death and intracellular α-synuclein (α-syn) aggregation. The interaction of iron accumulation with α-syn and tau was further explored as an essential pathological mechanism of PDD. However, the links and mechanisms between these factors remain unclear. Studies have shown that the occurrence and development of neurodegenerative diseases such as PDD are closely related to the separation of abnormal phases. Substances such as proteins can form droplets through liquid-liquid phase separation (LLPS) under normal physiological conditions and even undergo further liquid-solid phase transitions to form solid aggregates under disease or regulatory disorders, leading to pathological phenomena. By analyzing the existing literature, we propose that LLPS is the crucial mechanism causing abnormal accumulation of α-syn, tau, and other proteins in PDD, and its interaction with iron metabolism disorder is the key factor driving ferroptosis in PDD. Therefore, we believe that LLPS can serve as one of the means to explain the pathological mechanism of PDD. Determining the significance of LLPS in neurodegenerative diseases such as PDD will stimulate interest in research into treatments based on interference with abnormal LLPS.
Collapse
|
8
|
Tan B, Babur E, Toy N, Günaydın B, Dursun N, Süer C. Tau Protein is Differentially Phosphorylated in Young- and Old- Aged Rats with Experimentally Induced Hyperthyroidism. Int J Dev Neurosci 2022; 82:654-663. [PMID: 35904470 DOI: 10.1002/jdn.10220] [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: 03/29/2022] [Revised: 07/01/2022] [Accepted: 07/18/2022] [Indexed: 11/07/2022] Open
Abstract
AIM Aging involves progressive physiological changes, including thyroid dysfunction; thus, changes in plasma THs level may affect neuronal function such as synaptic plasticity and Tau phosphorylation. However, how tau protein is modulated in hyperthyroidism with aging is not clear. To clarify this issue, LTP and accompanying phosphorylation of Tau protein in different residues were investigated in the hippocampus of young and old rats with experimentally induced hyperthyroidism. MATERIALS AND METHODS The study was performed in vivo under urethane anesthesia on two-month and twelve-month-old Wistar albino male rats. Field potentials, composed of a field of excitatory-postsynaptic potential (fEPSP) and a population-spike (PS), occurring in the hippocampal dentate gyrus region, were recorded by applying high-frequency stimulation (HFS) to the perforant pathway (100 Hz, four times at five-minute intervals) to induce LTP. Total-and phosphorylated-tau were measured in HFS-induced hippocampus by using Western blotting. RESULTS The thyroid hormone suppressed hippocampal somatic LTP (PS) was suppressed with aging, and treatment improved this suppression in aged rats without any changes in synaptic LTP (fEPSP). The phosphorylation of Tau at Ser202/Thr205 and Thr231 residues was increased in aged control rats. Treatment of aged rats with L-Thyroxine reduced the phosphorylation of Tau at these residues to the young control condition. CONCLUSION Impaired LTP that occurs with aging may be among the underlying causes of dementia in relatively older ages, and L-Thyroxine treatment restores this impaired LTP. In addition, the phosphorylation level of Tau epitopes known to play a role in the pathogenesis of Alzheimer's disease may support a critical role in the modulation of synaptic plasticity in hyperthyroidism.
Collapse
Affiliation(s)
- Burak Tan
- Department of Physiology, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Ercan Babur
- Department of Physiology, School of Medicine, Tokat Gaziosmanpaşa University, Tokat, Turkey
| | - Nihal Toy
- Department of Physiology, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Buse Günaydın
- Department of Physiology, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Nurcan Dursun
- Department of Physiology, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Cem Süer
- Department of Physiology, School of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
9
|
Ferrer I, Andrés-Benito P, Garcia-Esparcia P, López-Gonzalez I, Valiente D, Jordán-Pirla M, Carmona M, Sala-Jarque J, Gil V, del Rio JA. Differences in Tau Seeding in Newborn and Adult Wild-Type Mice. Int J Mol Sci 2022; 23:4789. [PMID: 35563179 PMCID: PMC9099670 DOI: 10.3390/ijms23094789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 12/10/2022] Open
Abstract
Alzheimer's disease (AD) and other tauopathies are common neurodegenerative diseases in older adults; in contrast, abnormal tau deposition in neurons and glial cells occurs only exceptionally in children. Sarkosyl-insoluble fractions from sporadic AD (sAD) containing paired helical filaments (PHFs) were inoculated unilaterally into the thalamus in newborn and three-month-old wild-type C57BL/6 mice, which were killed at different intervals from 24 h to six months after inoculation. Tau-positive cells were scanty and practically disappeared at three months in mice inoculated at the age of a newborn. In contrast, large numbers of tau-positive cells, including neurons and oligodendrocytes, were found in the thalamus of mice inoculated at three months and killed at the ages of six months and nine months. Mice inoculated at the age of newborn and re-inoculated at the age of three months showed similar numbers and distribution of positive cells in the thalamus at six months and nine months. This study shows that (a) differences in tau seeding between newborn and young adults may be related to the ratios between 3Rtau and 4Rtau, and the shift to 4Rtau predominance in adults, together with the immaturity of connections in newborn mice, and (b) intracerebral inoculation of sAD PHFs in newborn mice does not protect from tau seeding following intracerebral inoculation of sAD PHFs in young/adult mice.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Pol Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Paula Garcia-Esparcia
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Irene López-Gonzalez
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Diego Valiente
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Mónica Jordán-Pirla
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Margarita Carmona
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Julia Sala-Jarque
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, Barcelona Institute for Science and Technology, Parc Científic de Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain; (J.S.-J.); (V.G.); (J.A.d.R.)
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, Barcelona Institute for Science and Technology, Parc Científic de Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain; (J.S.-J.); (V.G.); (J.A.d.R.)
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - José Antonio del Rio
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, Barcelona Institute for Science and Technology, Parc Científic de Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain; (J.S.-J.); (V.G.); (J.A.d.R.)
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
10
|
In vivo analysis of the phosphorylation of tau and the tau protein kinases Cdk5-p35 and GSK3β by using Phos-tag SDS–PAGE. J Proteomics 2022; 262:104591. [DOI: 10.1016/j.jprot.2022.104591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/24/2022]
|
11
|
Liang SY, Wang ZT, Tan L, Yu JT. Tau Toxicity in Neurodegeneration. Mol Neurobiol 2022; 59:3617-3634. [PMID: 35359226 DOI: 10.1007/s12035-022-02809-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/20/2022] [Indexed: 12/22/2022]
Abstract
Tau is a microtubule-associated protein widely distributed in the central nervous system (CNS). The main function of tau is to promote the assembly of microtubules and stabilize their structure. After a long period of research on neurodegenerative diseases, the function and dysfunction of the microtubule-associated protein tau in neurodegenerative diseases and tau neurotoxicity have attracted increasing attention. Tauopathies are a series of progressive neurodegenerative diseases caused by pathological changes in tau, such as abnormal phosphorylation. The pathological features of tauopathies are the deposition of abnormally phosphorylated tau proteins and the aggregation of tau proteins in neurons. This article first describes the normal physiological function and dysfunction of tau proteins and then discusses the enzymes and proteins involved in tau phosphorylation and dephosphorylation, the role of tau in cell dysfunction, and the relationships between tau and several neurodegenerative diseases. The study of tau neurotoxicity provides new directions for the treatment of tauopathies.
Collapse
Affiliation(s)
- Shu-Yu Liang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China. .,Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
12
|
Cryo-EM structures of τ filaments from human brain. Essays Biochem 2021; 65:949-959. [PMID: 34846514 PMCID: PMC8709893 DOI: 10.1042/ebc20210025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/01/2021] [Accepted: 10/14/2021] [Indexed: 12/30/2022]
Abstract
Electron cryo-microscopy (cryo-EM) has made it possible to determine near-atomic structures of τ filaments from human brain. Previous work had shown that the cores of paired helical and straight filaments of Alzheimer's disease are made of two identical, but differently arranged C-shaped protofilaments. In recent years, cryo-EM has shown that the Alzheimer τ fold is 79 amino acids long. Five of the eight β-strands give rise to two antiparallel β-sheets, with the other three forming a β-helix. High-affinity binding sites of positron emission tomography ligand APN-1607 (PM-PBB3) are in the β-helix region. The Alzheimer fold contrasts with the 94 amino acid-long Pick fold, which is J-shaped and comprises nine β-strands that give rise to four antiparallel β-sheets, in the absence of a β-helix. Chronic traumatic encephalopathy τ fold is similar to the Alzheimer fold, but differs in the β-helix region, which is larger and contains a non-proteinaceous density that is probably hydrophobic. These folds are mostly two-layered. By contrast, the 107 amino acid τ fold of the 4R tauopathy corticobasal degeneration is four-layered and comprises 11 β-strands. It contains an internal, probably hydrophilic, density that is surrounded by τ. The τ folds described here share the presence of microtubule-binding repeats 3 and 4, as well as 10-13 amino acids after repeat 4.
Collapse
|
13
|
Boyarko B, Hook V. Human Tau Isoforms and Proteolysis for Production of Toxic Tau Fragments in Neurodegeneration. Front Neurosci 2021; 15:702788. [PMID: 34744602 PMCID: PMC8566764 DOI: 10.3389/fnins.2021.702788] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/09/2021] [Indexed: 01/27/2023] Open
Abstract
The human tau protein is implicated in a wide range of neurodegenerative “tauopathy” diseases, consisting of Alzheimer’s disease (AD) and frontotemporal lobar degeneration which includes progressive supranuclear palsy, corticobasal degeneration, Pick’s disease, and FTLD-tau (frontotemporal dementia with parkinsonism caused by MAPT mutations). Tau gene transcripts in the human brain undergo alternative splicing to yield 6 different tau protein isoforms that are expressed in different ratios in neurodegeneration which result in tau pathology of paired-helical filaments, neurofibrillary tangles, and tau fibrillar aggregates with detrimental microtubule destabilization. Protease-mediated tau truncation is an important post-translational modification (PTM) which drives neurodegeneration in a tau fragment-dependent manner. While numerous tau fragments have been identified, knowledge of the proteolytic steps that convert each parent tau isoform into specific truncated tau fragments has not yet been fully defined. An improved understanding of the relationships between tau isoforms and their proteolytic processing to generate neurotoxic tau fragments is important to the field. This review evaluates tau isoform expression patterns including PTMs and mutations that influence proteolysis of tau to generate toxic fragments that drive cognitive deficits in AD and other tauopathy models. This assessment identifies the gap in the field on understanding the details of proteolytic steps used to convert each tau isoform into fragments. Knowledge of the processing mechanisms of tau isoforms can lead to new protease targeted drug strategies to prevent the formation of toxic tau fragments in tauopathy neurodegenerative diseases.
Collapse
Affiliation(s)
- Ben Boyarko
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States.,Department of Neurosciences and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
14
|
Samimi N, Sharma G, Kimura T, Matsubara T, Huo A, Chiba K, Saito Y, Murayama S, Akatsu H, Hashizume Y, Hasegawa M, Farjam M, Shahpasand K, Ando K, Hisanaga SI. Distinct phosphorylation profiles of tau in brains of patients with different tauopathies. Neurobiol Aging 2021; 108:72-79. [PMID: 34536819 DOI: 10.1016/j.neurobiolaging.2021.08.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/31/2021] [Accepted: 08/15/2021] [Indexed: 01/15/2023]
Abstract
Tauopathies are neurodegenerative diseases that are characterized by pathological accumulation of tau protein. Tau is hyperphosphorylated in the brain of tauopathy patients, and this phosphorylation is proposed to play a role in disease development. However, it has been unclear whether phosphorylation is different among different tauopathies. Here, we investigated the phosphorylation states of tau in several tauopathies, including corticobasal degeneration, Pick's disease, progressive supranuclear palsy (PSP), argyrophilic grain dementia (AGD) and Alzheimer's disease (AD). Analysis of tau phosphorylation profiles using Phos-tag SDS-PAGE revealed distinct phosphorylation of tau in different tauopathies, whereas similar phosphorylation patterns were found within the same tauopathy. For PSP, we found 2 distinct phosphorylation patterns suggesting that PSP may consist of 2 different related diseases. Immunoblotting with anti-phospho-specific antibodies showed different site-specific phosphorylation in the temporal lobes of patients with different tauopathies. AD brains showed increased phosphorylation at Ser202, Thr231 and Ser235, Pick's disease brains showed increased phospho-Ser202, and AGD brains showed increased phospho-Ser396. The cis conformation of the peptide bond between phospho-Thr231 and Pro232 (cis ptau) was increased in AD and AGD. These results indicate that while tau is differently phosphorylated in tauopathies, a similar pathological mechanism may occur in AGD and AD patients. The present data provide useful information regarding tau pathology and diagnosis of tauopathies.
Collapse
Affiliation(s)
- Nastaran Samimi
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Govinda Sharma
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Taeko Kimura
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Tomoyasu Matsubara
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan
| | - Anni Huo
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Kurumi Chiba
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Yuko Saito
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan
| | - Shigeo Murayama
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan
| | - Hiroyasu Akatsu
- Department of Community-based Medical Education, Nagoya City University Graduate School of Medicine, Mizuho, Nagoya, Aichi, Japan; Institute of Neuropathology, Fukushimura Hospital, Toyohashi, Aichi, Japan
| | - Yoshio Hashizume
- Institute of Neuropathology, Fukushimura Hospital, Toyohashi, Aichi, Japan
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan
| | - Mojtaba Farjam
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan; Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan.
| |
Collapse
|
15
|
Niu J, Iqbal K, Liu F, Hu W. Rats Display Sexual Dimorphism in Phosphorylation of Brain Tau with Age. J Alzheimers Dis 2021; 82:855-869. [PMID: 34092647 DOI: 10.3233/jad-210341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Women have a two-fold higher risk than men to Alzheimer's disease (AD) at midlife. Larger brain tau burden was consistently shown in older women than age-matched men. The biological basis for this gender disparity remains elusive. OBJECTIVE We sought to know whether tau expression and phosphorylation physiologically differ between males and females. METHODS We used western blots and immunohistochemistry to compare the levels of total tau and phosphorylated tau in the hippocampus and entorhinal cortex (EC) between sexes in Wistar rats at 40 days, and 8 and 20 months of age. RESULTS We detected no statistically significant difference in total tau, 3R-tau, and 4R-tau between sexes. However, female rats exhibited lower levels of tau unphosphorylated at the Tau-1 site at 40 days of age. At 8 months of age, females showed higher levels of tau phosphorylated at Ser190, Ser387, and Ser395 (Ser199, Ser396, and Ser404 of human tau, respectively) than males in EC. At 20 months of age, both brain regions of female rats consistently showed higher levels than males of tau phosphorylated at Ser253, Ser387, PHF-1 (Ser387/395), and Ser413 sites, which correspond to Ser262, Ser396, Ser396/404, and Ser422 of human tau, respectively. CONCLUSION Rats of both sexes have comparable levels of total tau, 3R-tau, and 4R-tau, whereas females exhibit higher levels of tau phosphorylated at multiple sites that are implicated in AD tau pathology, indicating a sexual dimorphism of tau phosphorylation that may potentially underlie the disparity in brain tau burden and risk for AD between sexes.
Collapse
Affiliation(s)
- Jiahui Niu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wen Hu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
16
|
García-Escudero V, Ruiz-Gabarre D, Gargini R, Pérez M, García E, Cuadros R, Hernández IH, Cabrera JR, García-Escudero R, Lucas JJ, Hernández F, Ávila J. A new non-aggregative splicing isoform of human Tau is decreased in Alzheimer's disease. Acta Neuropathol 2021; 142:159-177. [PMID: 33934221 PMCID: PMC8217066 DOI: 10.1007/s00401-021-02317-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Tauopathies, including Alzheimer's disease (AD) and frontotemporal lobar degeneration with Tau pathology (FTLD-tau), are a group of neurodegenerative disorders characterized by Tau hyperphosphorylation. Post-translational modifications of Tau such as phosphorylation and truncation have been demonstrated to be an essential step in the molecular pathogenesis of these tauopathies. In this work, we demonstrate the existence of a new, human-specific truncated form of Tau generated by intron 12 retention in human neuroblastoma cells and, to a higher extent, in human RNA brain samples, using qPCR and further confirming the results on a larger database of human RNA-seq samples. Diminished protein levels of this new Tau isoform are found by Westernblotting in Alzheimer's patients' brains (Braak I n = 3; Braak II n = 6, Braak III n = 3, Braak IV n = 1, and Braak V n = 10, Braak VI n = 8) with respect to non-demented control subjects (n = 9), suggesting that the lack of this truncated isoform may play an important role in the pathology. This new Tau isoform exhibits similar post-transcriptional modifications by phosphorylation and affinity for microtubule binding, but more interestingly, is less prone to aggregate than other Tau isoforms. Finally, we present evidence suggesting this new Tau isoform could be linked to the inhibition of GSK3β, which would mediate intron 12 retention by modulating the serine/arginine rich splicing factor 2 (SRSF2). Our results show the existence of an important new isoform of Tau and suggest that further research on this less aggregation-prone Tau may help to develop future therapies for Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Vega García-Escudero
- Departamento de Anatomía, Histología y Neurociencia, School of Medicine, Autonoma de Madrid University (UAM), Arzobispo Morcillo, 4, 28029, Madrid, Spain
- Graduate Program in Neuroscience, Autonoma de Madrid University (UAM), Arzobispo Morcillo, 4, 28029, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
| | - Daniel Ruiz-Gabarre
- Departamento de Anatomía, Histología y Neurociencia, School of Medicine, Autonoma de Madrid University (UAM), Arzobispo Morcillo, 4, 28029, Madrid, Spain
- Graduate Program in Neuroscience, Autonoma de Madrid University (UAM), Arzobispo Morcillo, 4, 28029, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
| | - Ricardo Gargini
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
- Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, 28220, Madrid, Spain
| | - Mar Pérez
- Departamento de Anatomía, Histología y Neurociencia, School of Medicine, Autonoma de Madrid University (UAM), Arzobispo Morcillo, 4, 28029, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
| | - Esther García
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
| | - Raquel Cuadros
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
| | - Ivó H Hernández
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
| | - Jorge R Cabrera
- Unidad de Investigación, Fundación Hospital de Jove, 33290, Gijón, Spain
| | - Ramón García-Escudero
- Molecular Oncology Unit, CIEMAT, Ave Complutense, 40, 28040, Madrid, Spain
- Hospital 12 Octubre Research Institute/CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), Valderrebollo, 5, 28031, Madrid, Spain
| | - José J Lucas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - Jesús Ávila
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM). Nicolás Cabrera, 1. Cantoblanco, 28049, Madrid, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031, Madrid, Spain.
| |
Collapse
|
17
|
4R Tau Modulates Cocaine-Associated Memory through Adult Dorsal Hippocampal Neurogenesis. J Neurosci 2021; 41:6753-6774. [PMID: 34099513 DOI: 10.1523/jneurosci.2848-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
The development, persistence and relapse of drug addiction require drug memory that generally develops with drug administration-paired contextual stimuli. Adult hippocampal neurogenesis (AHN) contributes to cocaine memory formation; however, the underlying mechanism remains unclear. Male mice hippocampal expression of Tau was significantly decreased during the cocaine-associated memory formation. Genetic overexpression of four microtubule-binding repeats Tau (4R Tau) in the mice hippocampus disrupted cocaine memory by suppressing AHN. Furthermore, 4R Tau directly interacted with phosphoinositide 3-kinase (PI3K)-p85 and impaired its nuclear translocation and PI3K-AKT signaling, processes required for hippocampal neuron proliferation. Collectively, 4R Tau modulates cocaine memory formation by disrupting AHN, suggesting a novel mechanism underlying cocaine memory formation and provide a new strategy for the treatment of cocaine addiction.SIGNIFICANCE STATEMENT Drug memory that generally develops with drug-paired contextual stimuli and drug administration is critical for the development, persistence and relapse of drug addiction. Previous studies have suggested that adult hippocampal neurogenesis (AHN) plays a role in cocaine memory formation. Here, we showed that Tau was significantly downregulated in the hippocampus in the cocaine memory formation. Tau knock-out (KO) promoted AHN in the hippocampal dentate gyrus (DG), resulting in the enhanced memory formation evoked by cocaine-cue stimuli. In contrast, genetically overexpressed 4R Tau in the hippocampus disrupted cocaine-cue memory by suppressing AHN. In addition, 4R Tau interacted directly with phosphoinositide 3-kinase (PI3K)-p85 and hindered its nuclear translocation, eventually repressing PI3K-AKT signaling, which is essential for hippocampal neuronal proliferation.
Collapse
|
18
|
Candelise N, Scaricamazza S, Salvatori I, Ferri A, Valle C, Manganelli V, Garofalo T, Sorice M, Misasi R. Protein Aggregation Landscape in Neurodegenerative Diseases: Clinical Relevance and Future Applications. Int J Mol Sci 2021; 22:ijms22116016. [PMID: 34199513 PMCID: PMC8199687 DOI: 10.3390/ijms22116016] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/13/2022] Open
Abstract
Intrinsic disorder is a natural feature of polypeptide chains, resulting in the lack of a defined three-dimensional structure. Conformational changes in intrinsically disordered regions of a protein lead to unstable β-sheet enriched intermediates, which are stabilized by intermolecular interactions with other β-sheet enriched molecules, producing stable proteinaceous aggregates. Upon misfolding, several pathways may be undertaken depending on the composition of the amino acidic string and the surrounding environment, leading to different structures. Accumulating evidence is suggesting that the conformational state of a protein may initiate signalling pathways involved both in pathology and physiology. In this review, we will summarize the heterogeneity of structures that are produced from intrinsically disordered protein domains and highlight the routes that lead to the formation of physiological liquid droplets as well as pathogenic aggregates. The most common proteins found in aggregates in neurodegenerative diseases and their structural variability will be addressed. We will further evaluate the clinical relevance and future applications of the study of the structural heterogeneity of protein aggregates, which may aid the understanding of the phenotypic diversity observed in neurodegenerative disorders.
Collapse
Affiliation(s)
- Niccolò Candelise
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.); (A.F.); (C.V.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-338-891-2668
| | - Silvia Scaricamazza
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.); (A.F.); (C.V.)
| | - Illari Salvatori
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.); (A.F.); (C.V.)
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy; (V.M.); (T.G.); (M.S.); (R.M.)
| | - Alberto Ferri
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.); (A.F.); (C.V.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Cristiana Valle
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.); (A.F.); (C.V.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Valeria Manganelli
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy; (V.M.); (T.G.); (M.S.); (R.M.)
| | - Tina Garofalo
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy; (V.M.); (T.G.); (M.S.); (R.M.)
| | - Maurizio Sorice
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy; (V.M.); (T.G.); (M.S.); (R.M.)
| | - Roberta Misasi
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy; (V.M.); (T.G.); (M.S.); (R.M.)
| |
Collapse
|
19
|
Habekost M, Qvist P, Denham M, Holm IE, Jørgensen AL. Directly Reprogrammed Neurons Express MAPT and APP Splice Variants Pertinent to Ageing and Neurodegeneration. Mol Neurobiol 2021; 58:2075-2087. [PMID: 33415685 PMCID: PMC8018937 DOI: 10.1007/s12035-020-02258-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Neurons produced by reprogramming of other cell types are used to study cellular mechanisms of age-related neurodegenerative diseases. To model Alzheimer's disease and other tauopathies, it is essential that alternative splicing of the MAPT transcript in these neurons produces the relevant tau isoforms. Human neurons derived from induced pluripotent stem cells, however, express tau isoform compositions characteristic of foetal neurons rather than of adult neurons unless cultured in vitro for extended time periods. In this study, we characterised the dynamics of the MAPT and APP alternative splicing during a developmental time-course of porcine and murine cerebral cortices. We found age-dependent and species-specific isoform composition of MAPT, including 3R and 4R isoforms in the porcine adult brain similar to that of the adult human brain. We converted adult and embryonic fibroblasts directly into induced neurons and found similar developmental patterns of isoform composition, notably, the 3R and 4R isoforms relevant to the pathogenesis of Alzheimer's disease. Also, we observed cell-type-specific isoform expression of APP transcripts during the conversion. The approach was further used to generate induced neurons from transgenic pigs carrying Alzheimer's disease-causing mutations. We show that such neurons authentically model the first crucial steps in AD pathogenesis.
Collapse
Affiliation(s)
- Mette Habekost
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark.
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark.
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- iPSYCH, Lundbeck Foundation Initiative for Integrative Psychiatric Research, 8000C, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, 8000C, Aarhus, Denmark
| | - Mark Denham
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Ida E Holm
- Department of Pathology, Randers Hospital, 8930, Randers, Denmark
- Department of Clinical Medicine, Aarhus University, 8000C, Aarhus, Denmark
| | | |
Collapse
|
20
|
Wei Y, Liu M, Wang D. The propagation mechanisms of extracellular tau in Alzheimer's disease. J Neurol 2021; 269:1164-1181. [PMID: 33913022 DOI: 10.1007/s00415-021-10573-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 01/07/2023]
Abstract
Tubulin-associated unit (tau) is an important microtubule-associated protein. The abnormal intracellular aggregation of tau has been strongly associated with Alzheimer's disease (AD). Accumulating evidence has conclusively demonstrated that tau is present in the cytoplasm of neurons and is also actively released into the extracellular space. However, the types of tau species that are released are unclear, as is the mechanism of their release by donor neurons and subsequent uptake by recipient neurons in AD. Understanding the underlying mechanisms of abnormal tau cell-to-cell transmission can provide novel insights into the etiology and pathogenesis of AD and can help identify new targets for the development of AD therapies focused on counteracting neurodegeneration or even preventing it. From this perspective, the present review focuses on recent advances in understanding the mechanisms regulating the levels of extracellular tau and discusses the role of such mechanisms in the propagation of tau-associated pathology.
Collapse
Affiliation(s)
- Yun Wei
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing city, 100091, China.
| | - Meixia Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing city, 100091, China
| | - Dongxin Wang
- Jining Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shandong province, 272000, China
| |
Collapse
|
21
|
Morris SL, Tsai MY, Aloe S, Bechberger K, König S, Morfini G, Brady ST. Defined Tau Phosphospecies Differentially Inhibit Fast Axonal Transport Through Activation of Two Independent Signaling Pathways. Front Mol Neurosci 2021; 13:610037. [PMID: 33568975 PMCID: PMC7868336 DOI: 10.3389/fnmol.2020.610037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/28/2020] [Indexed: 12/30/2022] Open
Abstract
Tau protein is subject to phosphorylation by multiple kinases at more than 80 different sites. Some of these sites are associated with tau pathology and neurodegeneration, but other sites are modified in normal tau as well as in pathological tau. Although phosphorylation of tau at residues in the microtubule-binding repeats is thought to reduce tau association with microtubules, the functional consequences of other sites are poorly understood. The AT8 antibody recognizes a complex phosphoepitope site on tau that is detectable in a healthy brain but significantly increased in Alzheimer's disease (AD) and other tauopathies. Previous studies showed that phosphorylation of tau at the AT8 site leads to exposure of an N-terminal sequence that promotes activation of a protein phosphatase 1 (PP1)/glycogen synthase 3 (GSK3) signaling pathway, which inhibits kinesin-1-based anterograde fast axonal transport (FAT). This finding suggests that phosphorylation may control tau conformation and function. However, the AT8 includes three distinct phosphorylated amino acids that may be differentially phosphorylated in normal and disease conditions. To evaluate the effects of specific phosphorylation sites in the AT8 epitope, recombinant, pseudophosphorylated tau proteins were perfused into the isolated squid axoplasm preparation to determine their effects on axonal signaling pathways and FAT. Results from these studies suggest a mechanism where specific phosphorylation events differentially impact tau conformation, promoting activation of independent signaling pathways that differentially affect FAT. Implications of findings here to our understanding of tau function in health and disease conditions are discussed.
Collapse
Affiliation(s)
- Sarah L. Morris
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- Marine Biological Laboratory, Woods Hole, MA, United States
| | - Ming-Ying Tsai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Sarah Aloe
- Marine Biological Laboratory, Woods Hole, MA, United States
| | | | - Svenja König
- Marine Biological Laboratory, Woods Hole, MA, United States
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- Marine Biological Laboratory, Woods Hole, MA, United States
| | - Scott T. Brady
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- Marine Biological Laboratory, Woods Hole, MA, United States
| |
Collapse
|
22
|
Hadi F, Akrami H, Totonchi M, Barzegar A, Nabavi SM, Shahpasand K. α-synuclein abnormalities trigger focal tau pathology, spreading to various brain areas in Parkinson disease. J Neurochem 2021; 157:727-751. [PMID: 33264426 DOI: 10.1111/jnc.15257] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/28/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder, whose prevalence is 2~3% in the population over 65. α-Synuclein aggregation is the major pathological hallmark of PD. However, recent studies have demonstrated enhancing evidence of tau pathology in PD. Despite extensive considerations, thus far, the actual spreading mechanism of neurodegeneration has remained elusive in a PD brain. This study aimed to further investigate the development of α-synuclein and tau pathology. We employed various PD models, including cultured neurons treated with either 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or with recombinant α-synuclein. Also, we studied dopaminergic neurons of cytokine Interferon-β knock-out. Moreover, we examined rats treated with 6-hydroxydopamine, Rhesus monkeys administrated with MPTP neurotoxin, and finally, human post-mortem brains. We found the α-synuclein phosphorylation triggers tau pathogenicity. Also, we observed more widespread phosphorylated tau than α-synuclein with prion-like nature in various brain areas. We optionally removed P-tau or P-α-synuclein from cytokine interferon-β knock out with respective monoclonal antibodies. We found that tau immunotherapy suppressed neurodegeneration more than α-synuclein elimination. Our findings indicate that the pathogenic tau could be one of the leading causes of comprehensive neurodegeneration triggered by PD. Thus, we can propose an efficient therapeutic target to fight the devastating disorder.
Collapse
Affiliation(s)
- Fatemeh Hadi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Hassan Akrami
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Mehdi Totonchi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | | | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| |
Collapse
|
23
|
Duquette A, Pernègre C, Veilleux Carpentier A, Leclerc N. Similarities and Differences in the Pattern of Tau Hyperphosphorylation in Physiological and Pathological Conditions: Impacts on the Elaboration of Therapies to Prevent Tau Pathology. Front Neurol 2021; 11:607680. [PMID: 33488502 PMCID: PMC7817657 DOI: 10.3389/fneur.2020.607680] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
Tau protein, a neuronal microtubule-associated protein, becomes hyperphosphorylated in several neurodegenerative diseases called tauopathies. Hyperphosphorylation of tau is correlated to its redistribution from the axon to the somato-dendritic compartment at early stages of tauopathies. Interestingly, tau hyperphosphorylation begins in different regions of the brain in each tauopathy. In some regions, both neurons and glial cells develop tau hyperphosphorylation. Tau hyperphosphorylation is also observed in physiological conditions such as hibernation and brain development. In the first section of present article, we will review the spatiotemporal and cellular distribution of hyperphosphorylated tau in the most frequent tauopathies. In the second section, we will compare the pattern of tau hyperphosphorylation in physiological and pathological conditions and discuss the sites that could play a pivotal role in the conversion of non-toxic to toxic forms of hyperphosphorylated tau. Furthermore, we will discuss the role of hyperphosphorylated tau in physiological and pathological conditions and the fact that tau hyperphosphorylation is reversible in physiological conditions but not in a pathological ones. In the third section, we will speculate how the differences and similarities between hyperphosphorylated tau in physiological and pathological conditions could impact the elaboration of therapies to prevent tau pathology. In the fourth section, the different therapeutic approaches using tau as a direct or indirect therapeutic target will be presented.
Collapse
Affiliation(s)
- Antoine Duquette
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Camille Pernègre
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Ariane Veilleux Carpentier
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Nicole Leclerc
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
24
|
Ishunina TA. Alternative splicing in aging and Alzheimer's disease: Highlighting the role of tau and estrogen receptor α isoforms in the hypothalamus. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:177-189. [PMID: 34266591 DOI: 10.1016/b978-0-12-819973-2.00012-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human genes show the highest efficacy of alternative splicing (AS) in the brain as compared to other tissues. Within the brain, a remarkably rich diversity of AS events was identified in the hypothalamus. The AS frequency is increased in the aging brain. Such AS events, as intron retention and accumulation of circular RNAs, were acknowledged as some of the main hallmarks of the aging brain. In Alzheimer's disease (AD) pivotal (tau gene, in particular), risk, candidate and other genes show significant alterations in AS. Therefore AD has been suggested to be a disease of dysregulated AS. One of the reported risk factors for AD is estrogen deficiency that may interfere with the extension of neurobrillary tangles. Mounting evidence suggests that estrogens may decrease hyperphosphorylated tau deposition in the brain. Furthermore, AS of estrogen receptor α (ERα) mRNA is decreased in AD brain areas with the highest tau load. These potential interactions among tau, estrogens, and ERα AS may be important for the development of therapeutic and preventive strategies for AD. The intriguing point is that the amount of splice variants of ERα in the hypothalamus and the hippocampus is increased in aging and decreased in AD, while ERα is one of the regulators of AS and is subject to AS itself.
Collapse
Affiliation(s)
- Tatjana A Ishunina
- Department of Histology, Embryology and Cytology, Kursk State Medical University, Kursk, Russia.
| |
Collapse
|
25
|
Goedert M, Spillantini MG, Falcon B, Zhang W, Newell KL, Hasegawa M, Scheres SHW, Ghetti B. Tau Protein and Frontotemporal Dementias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:177-199. [PMID: 33433876 DOI: 10.1007/978-3-030-51140-1_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Filamentous inclusions of tau protein are found in cases of inherited and sporadic frontotemporal dementias (FTDs). Mutations in MAPT, the tau gene, cause approximately 5% of cases of FTD. They proved that dysfunction of tau protein is sufficient to cause neurodegeneration and dementia. Clinically and pathologically, cases with MAPT mutations can resemble sporadic diseases, such as Pick's disease, globular glial tauopathy, progressive supranuclear palsy and corticobasal degeneration. The structures of tau filaments from Pick's disease and corticobasal degeneration, determined by electron cryo-microscopy, revealed the presence of specific tau folds in each disease, with no inter-individual variation. The same was true of chronic traumatic encephalopathy.
Collapse
Affiliation(s)
| | | | | | | | - Kathy L Newell
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN, USA
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
26
|
Tau modulates visual plasticity in adult and old mice. Neurobiol Aging 2020; 95:214-224. [DOI: 10.1016/j.neurobiolaging.2020.07.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/10/2020] [Accepted: 07/25/2020] [Indexed: 11/20/2022]
|
27
|
Goedert M. Tau proteinopathies and the prion concept. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:239-259. [PMID: 32958235 DOI: 10.1016/bs.pmbts.2020.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ordered assembly of a small number of proteins into amyloid filaments is central to age-related neurodegenerative diseases. Tau is the most commonly affected of these proteins. In sporadic diseases, assemblies of tau form in a stochastic manner in certain brain regions, from where they appear to spread in a deterministic way, giving rise to disease symptoms. Over the past decade, multiple lines of evidence have shown that assembled tau behaves like a prion. More recently, electron cryo-microscopy of tau filaments has shown that distinct conformers are present in different diseases, with no inter-individual variation for a given disease.
Collapse
Affiliation(s)
- Michel Goedert
- MRC Laboratory Molecular Biology, Cambridge, United Kingdom.
| |
Collapse
|
28
|
Köppen J, Schulze A, Machner L, Wermann M, Eichentopf R, Guthardt M, Hähnel A, Klehm J, Kriegeskorte MC, Hartlage-Rübsamen M, Morawski M, von Hörsten S, Demuth HU, Roßner S, Schilling S. Amyloid-Beta Peptides Trigger Aggregation of Alpha-Synuclein In Vitro. Molecules 2020; 25:E580. [PMID: 32013170 PMCID: PMC7037551 DOI: 10.3390/molecules25030580] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD), including dementia with Lewy bodies (DLB), account for the majority of dementia cases worldwide. Interestingly, a significant number of patients have clinical and neuropathological features of both AD and PD, i.e., the presence of amyloid deposits and Lewy bodies in the neocortex. The identification of α-synuclein peptides in amyloid plaques in DLB brain led to the hypothesis that both peptides mutually interact with each other to facilitate neurodegeneration. In this article, we report the influence of Aβ(1-42) and pGlu-Aβ(3-42) on the aggregation of α-synuclein in vitro. The aggregation of human recombinant α-synuclein was investigated using thioflavin-T fluorescence assay. Fibrils were investigated by means of antibody conjugated immunogold followed by transmission electron microscopy (TEM). Our data demonstrate a significantly increased aggregation propensity of α-synuclein in the presence of minor concentrations of Aβ(1-42) and pGlu-Aβ(3-42) for the first time, but without effect on toxicity on mouse primary neurons. The analysis of the composition of the fibrils by TEM combined with immunogold labeling of the peptides revealed an interaction of α-synuclein and Aβ in vitro, leading to an accelerated fibril formation. The analysis of kinetic data suggests that significantly enhanced nucleus formation accounts for this effect. Additionally, co-occurrence of α-synuclein and Aβ and pGlu-Aβ, respectively, under pathological conditions was confirmed in vivo by double immunofluorescent labelings in brains of aged transgenic mice with amyloid pathology. These observations imply a cross-talk of the amyloid peptides α-synuclein and Aβ species in neurodegeneration. Such effects might be responsible for the co-occurrence of Lewy bodies and plaques in many dementia cases.
Collapse
Affiliation(s)
- Janett Köppen
- Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation IZI-MWT, 06120 Halle, Germany; (J.K.); (L.M.); (M.W.); (R.E.); (H.-U.D.); (S.S.)
| | - Anja Schulze
- Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation IZI-MWT, 06120 Halle, Germany; (J.K.); (L.M.); (M.W.); (R.E.); (H.-U.D.); (S.S.)
| | - Lisa Machner
- Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation IZI-MWT, 06120 Halle, Germany; (J.K.); (L.M.); (M.W.); (R.E.); (H.-U.D.); (S.S.)
| | - Michael Wermann
- Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation IZI-MWT, 06120 Halle, Germany; (J.K.); (L.M.); (M.W.); (R.E.); (H.-U.D.); (S.S.)
| | - Rico Eichentopf
- Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation IZI-MWT, 06120 Halle, Germany; (J.K.); (L.M.); (M.W.); (R.E.); (H.-U.D.); (S.S.)
| | - Max Guthardt
- Fraunhofer Institute of Cell Therapy and Immunology IZI, 04103 Leipzig, Germany;
| | - Angelika Hähnel
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, 06120 Halle, Germany; (A.H.); (J.K.)
| | - Jessica Klehm
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, 06120 Halle, Germany; (A.H.); (J.K.)
| | - Marie-Christin Kriegeskorte
- Paul Flechsig Institute of Brain Research, University of Leipzig, 04109 Leipzig, Germany; (M.-C.K.); (M.H.-R.); (M.M.); (S.R.)
| | - Maike Hartlage-Rübsamen
- Paul Flechsig Institute of Brain Research, University of Leipzig, 04109 Leipzig, Germany; (M.-C.K.); (M.H.-R.); (M.M.); (S.R.)
| | - Markus Morawski
- Paul Flechsig Institute of Brain Research, University of Leipzig, 04109 Leipzig, Germany; (M.-C.K.); (M.H.-R.); (M.M.); (S.R.)
| | - Stephan von Hörsten
- Friedrich-Alexander-University Erlangen-Nürnberg, Preclinical Experimental Center, 91054 Erlangen, Germany;
| | - Hans-Ulrich Demuth
- Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation IZI-MWT, 06120 Halle, Germany; (J.K.); (L.M.); (M.W.); (R.E.); (H.-U.D.); (S.S.)
| | - Steffen Roßner
- Paul Flechsig Institute of Brain Research, University of Leipzig, 04109 Leipzig, Germany; (M.-C.K.); (M.H.-R.); (M.M.); (S.R.)
| | - Stephan Schilling
- Fraunhofer Institute of Cell Therapy and Immunology, Department of Drug Design and Target Validation IZI-MWT, 06120 Halle, Germany; (J.K.); (L.M.); (M.W.); (R.E.); (H.-U.D.); (S.S.)
| |
Collapse
|
29
|
Hefti MM, Kim S, Bell AJ, Betters RK, Fiock KL, Iida MA, Smalley ME, Farrell K, Fowkes ME, Crary JF. Tau Phosphorylation and Aggregation in the Developing Human Brain. J Neuropathol Exp Neurol 2019; 78:930-938. [PMID: 31504677 PMCID: PMC6751069 DOI: 10.1093/jnen/nlz073] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Tau hyperphosphorylation, mostly at serine (Ser) or threonine (Thr) residues, plays a key role in the pathogenesis of Alzheimer disease (AD) and other tauopathies. Rodent studies show similar hyperphosphorylation in the developing brain, which may be involved in regulating axonal growth and plasticity, but detailed human studies are lacking. Here, we examine tau phosphorylation by immunohistochemistry and immunoblotting in human fetal and adult autopsy brain tissue. Of the 20 cases with sufficient tissue preservation, 18 (90%) showed positive staining for S214 (pSer214), with the majority also positive for CP13 (pSer202), and PHF-1 (pSer396/pSer404). AT8 (pSer202/pThr205) and RZ3 (pThr231) were largely negative while PG5 (pSer409) was negative in all cases. Immunoblotting showed tau monomers with a similar staining pattern. We also observed phospho-tau aggregates in the fetal molecular layer, staining positively for S214, CP13, and PHF1 and negative for thioflavin S. These corresponded to high-molecular weight (∼150 kD) bands seen on Western blots probed with S214, PHF1, and PG5. We therefore conclude that fetal phosphorylation overlaps with AD in some residues, while others (e.g. T231, S409) appear to be unique to AD, and that tau is capable of forming nontoxic aggregates in the developing brain. These findings suggest that the fetal brain is resilient to formation of toxic aggregates, the mechanism for which may yield insights into the pathogenesis of tau aggregation and toxicity in the aging brain.
Collapse
Affiliation(s)
- Marco M Hefti
- Department of Pathology
- Iowa Neuroscience Institute
- Interdisciplinary Neuroscience Graduate Program, University of Iowa, Iowa City, Iowa
| | - SoongHo Kim
- Iowa Neuroscience Institute
- Department of Neuroscience
- Friedmann Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aaron J Bell
- Iowa Neuroscience Institute
- Analytical Instrumentation Facility Campus Box 7531, Monteith Research Center, 2410 Campus Shore Drive, Room 246, Raleigh, NC 27695-7531
| | - Ryan K Betters
- Interdisciplinary Neuroscience Graduate Program, University of Iowa, Iowa City, Iowa
| | | | - Megan A Iida
- Iowa Neuroscience Institute
- Department of Neuroscience
- Friedmann Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Kurt Farrell
- Iowa Neuroscience Institute
- Department of Neuroscience
- Friedmann Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - John F Crary
- Iowa Neuroscience Institute
- Department of Neuroscience
- Friedmann Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
30
|
Sharma G, Huo A, Kimura T, Shiozawa S, Kobayashi R, Sahara N, Ishibashi M, Ishigaki S, Saito T, Ando K, Murayama S, Hasegawa M, Sobue G, Okano H, Hisanaga SI. Tau isoform expression and phosphorylation in marmoset brains. J Biol Chem 2019; 294:11433-11444. [PMID: 31171723 DOI: 10.1074/jbc.ra119.008415] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/23/2019] [Indexed: 01/07/2023] Open
Abstract
Tau is a microtubule-associated protein expressed in neuronal axons. Hyperphosphorylated tau is a major component of neurofibrillary tangles, a pathological hallmark of Alzheimer's disease (AD). Hyperphosphorylated tau aggregates are also found in many neurodegenerative diseases, collectively referred to as "tauopathies," and tau mutations are associated with familial frontotemporal lobar degeneration (FTLD). Previous studies have generated transgenic mice with mutant tau as tauopathy models, but nonhuman primates, which are more similar to humans, may be a better model to study tauopathies. For example, the common marmoset is poised as a nonhuman primate model for investigating the etiology of age-related neurodegenerative diseases. However, no biochemical studies of tau have been conducted in marmoset brains. Here, we investigated several important aspects of tau, including expression of different tau isoforms and its phosphorylation status, in the marmoset brain. We found that marmoset tau does not possess the "primate-unique motif" in its N-terminal domain. We also discovered that the tau isoform expression pattern in marmosets is more similar to that of mice than that of humans, with adult marmoset brains expressing only four-repeat tau isoforms as in adult mice but unlike in adult human brains. Of note, tau in brains of marmoset newborns was phosphorylated at several sites associated with AD pathology. However, in adult marmoset brains, much of this phosphorylation was lost, except for Ser-202 and Ser-404 phosphorylation. These results reveal key features of tau expression and phosphorylation in the marmoset brain, a potentially useful nonhuman primate model of neurodegenerative diseases.
Collapse
Affiliation(s)
- Govinda Sharma
- Laboratory of Molecular Neuroscience, Faculty of Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Anni Huo
- Laboratory of Molecular Neuroscience, Faculty of Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Taeko Kimura
- Laboratory of Molecular Neuroscience, Faculty of Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan.,Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage, Chiba, Chiba 263-8555, Japan
| | - Seiji Shiozawa
- Department of Physiology, School of Medicine, Keio University, Shinjuku, Tokyo, 160-8582, Japan
| | - Reona Kobayashi
- Department of Physiology, School of Medicine, Keio University, Shinjuku, Tokyo, 160-8582, Japan.,Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage, Chiba, Chiba 263-8555, Japan
| | - Minaka Ishibashi
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Shinsuke Ishigaki
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Taro Saito
- Laboratory of Molecular Neuroscience, Faculty of Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Kanae Ando
- Laboratory of Molecular Neuroscience, Faculty of Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Shigeo Murayama
- Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Masato Hasegawa
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| | - Gen Sobue
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Shinjuku, Tokyo, 160-8582, Japan.,Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Shin-Ichi Hisanaga
- Laboratory of Molecular Neuroscience, Faculty of Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan .,Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| |
Collapse
|
31
|
Vasili E, Dominguez-Meijide A, Outeiro TF. Spreading of α-Synuclein and Tau: A Systematic Comparison of the Mechanisms Involved. Front Mol Neurosci 2019; 12:107. [PMID: 31105524 PMCID: PMC6494944 DOI: 10.3389/fnmol.2019.00107] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 04/09/2019] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are age-associated neurodegenerative disorders characterized by the misfolding and aggregation of alpha-synuclein (aSyn) and tau, respectively. The coexistence of aSyn and tau aggregates suggests a strong overlap between tauopathies and synucleinopathies. Interestingly, misfolded forms of aSyn and tau can propagate from cell to cell, and throughout the brain, thereby templating the misfolding of native forms of the proteins. The exact mechanisms involved in the propagation of the two proteins show similarities, and are reminiscent of the spreading characteristic of prion diseases. Recently, several models were developed to study the spreading of aSyn and tau. Here, we discuss the mechanisms involved, the similarities and differences between the spreading of the two proteins and that of the prion protein, and the different cell and animal models used for studying these processes. Ultimately, a deeper understanding of the molecular mechanisms involved may lead to the identification of novel targets for therapeutic intervention in a variety of devastating neurodegenerative diseases.
Collapse
Affiliation(s)
- Eftychia Vasili
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Antonio Dominguez-Meijide
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany.,Max Planck Institute for Experimental Medicine, Goettingen, Germany.,The Medical School, Institute of Neuroscience, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
32
|
Goedert M, Falcon B, Zhang W, Ghetti B, Scheres SHW. Distinct Conformers of Assembled Tau in Alzheimer's and Pick's Diseases. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:163-171. [PMID: 30886056 DOI: 10.1101/sqb.2018.83.037580] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tau filaments with distinct morphologies and/or isoform compositions underlie a large number of human neurodegenerative diseases. In conjunction with experimental studies, this has led to the suggestion that conformers of aggregated tau exist. Electron cryo-microscopy can be used to determine high-resolution structures of amyloid filaments from human brain. Paired helical and straight tau filaments of Alzheimer's disease (AD) are ultrastructural polymorphs. Each filament core is composed of two identical protofilaments extending from G273/304-E380 (in the numbering of the 441-amino acid isoform of human tau), which adopt a combined cross-β/β-helix structure. They comprise the ends of the first or second microtubule-binding repeat (R1 or R2), the whole of R3 and R4, and 12 amino acids after R4. In contrast, the core of the narrow filaments of Pick's disease (PiD) consists of a single protofilament extending from K254-F378 of 3R tau, which adopts a cross-β structure. It comprises the last 21 amino acids of R1, all of R3 and R4, and 10 amino acids after R4. Wide tau filaments of PiD, which are in the minority, consist of two narrow filaments packed against each other. The tau filament folds of AD and PiD appear to be conserved between different cases of disease. These findings show that filamentous tau adopts one fold in AD and a different fold in PiD, establishing the existence of distinct conformers.
Collapse
Affiliation(s)
- Michel Goedert
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Benjamin Falcon
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Wenjuan Zhang
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Sjors H W Scheres
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
33
|
Ordered Assembly of Tau Protein and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:3-21. [PMID: 32096024 DOI: 10.1007/978-981-32-9358-8_1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Tau filaments with distinct morphologies and/or isoform compositions underlie a large number of human neurodegenerative diseases. Their formation is important, because dominantly inherited mutations in MAPT, the tau gene, cause frontotemporal dementia with abundant filamentous tau inclusions. Assembly of tau may begin in a specific region of the brain, from where it spreads to other areas. It remains to be seen if the molecular species underlying tau aggregate-mediated neurodegeneration and propagation are the same or different. In the brains of mice transgenic for human mutant P301S tau, small tau filaments are the predominant seed-competent species. It has been suggested that different conformers of assembled tau may give rise to different human tauopathies, but until recently, it was not possible to study this directly. Electron cryo-microscopy can now be used to determine high-resolution structures of amyloid filaments from human brain. Paired helical and straight tau filaments of Alzheimer's disease are ultrastructural polymorphs. Each filament core is composed of two identical protofilaments extending from G273/304-E380 (in the numbering of the 441 amino acid isoform of human tau), which adopt a combined cross-β/β-helix structure. They comprise the ends of the first or second microtubule-binding repeat (R1 or R2), the whole of R3 and R4, as well as 12 amino acids after R4. By contrast, the core of the narrow filament of Pick's disease consists of a single protofilament extending from K254-F378 of 3R tau, which adopts a cross-β structure. It comprises the last 21 amino acids of R1, all of R3 and R4, as well as 10 amino acids after R4. Wide tau filaments of Pick's disease, which are in the minority, consist of two narrow filaments packed against each other. The tau filament folds of Alzheimer's and Pick's diseases appear to be conserved between different cases of disease. These findings show that filamentous tau adopts one fold in Alzheimer's disease and a different fold in Pick's disease, establishing the existence of distinct conformers.
Collapse
|
34
|
Goedert M. Tau filaments in neurodegenerative diseases. FEBS Lett 2018; 592:2383-2391. [PMID: 29790176 DOI: 10.1002/1873-3468.13108] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 11/11/2022]
Abstract
The ordered assembly of Tau protein into abnormal filamentous inclusions is a defining characteristic of many human neurodegenerative diseases. Thirty years ago, we reported that Tau is an integral component of the intraneuronal filaments of Alzheimer's disease. All six brain Tau isoforms make up those filaments. Twenty years ago, we and others showed that mutations in MAPT, the Tau gene, cause familial forms of frontotemporal dementia, thus proving that dysfunction of Tau protein is sufficient to cause neurodegeneration and dementia. More recently, we showed that high-resolution structures of Tau filaments from human brain can be determined by electron cryo-microscopy. These filaments may form the seeds that underlie the prion-like properties of aggregated tau.
Collapse
|
35
|
Goedert M, Yamaguchi Y, Mishra SK, Higuchi M, Sahara N. Tau Filaments and the Development of Positron Emission Tomography Tracers. Front Neurol 2018; 9:70. [PMID: 29497399 PMCID: PMC5818396 DOI: 10.3389/fneur.2018.00070] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/30/2018] [Indexed: 12/15/2022] Open
Abstract
A pathological pathway leading from soluble, monomeric to insoluble, filamentous Tau, is believed to underlie human Tauopathies. Cases of frontotemporal dementia are caused by dominantly inherited mutations in MAPT, the Tau gene. They show that dysfunction of Tau protein is sufficient to cause neurodegeneration and dementia. Extrapolation to the more common sporadic Tauopathies leads one to conclude that the pathological pathway is central to the development of all cases of disease, even if there are multiple reasons for Tau assembly. These findings are conceptually similar to those reported for beta-amyloid, alpha-synuclein and prion protein. Here, we provide an overview of Tau filaments and their positron emission tomography ligands.
Collapse
Affiliation(s)
- Michel Goedert
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba, Japan
| | | |
Collapse
|