1
|
Wiesner D, Feldengut S, Woelfle S, Boeckers TM, Ludolph AC, Roselli F, Del Tredici K. Neuropeptide FF (NPFF)-positive nerve cells of the human cerebral cortex and white matter in controls, selected neurodegenerative diseases, and schizophrenia. Acta Neuropathol Commun 2024; 12:108. [PMID: 38943180 PMCID: PMC11212262 DOI: 10.1186/s40478-024-01792-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/02/2024] [Indexed: 07/01/2024] Open
Abstract
We quantified and determined for the first time the distribution pattern of the neuropeptide NPFF in the human cerebral cortex and subjacent white matter. To do so, we studied n = 9 cases without neurological disorders and n = 22 cases with neurodegenerative diseases, including sporadic amyotrophic lateral sclerosis (ALS, n = 8), Alzheimer's disease (AD, n = 8), Pick's disease (PiD, n = 3), and schizophrenia (n = 3). NPFF-immunopositive cells were located chiefly, but not exclusively, in the superficial white matter and constituted there a subpopulation of white matter interstitial cells (WMIC): Pyramidal-like and multipolar somata predominated in the gyral crowns, whereas bipolar and ovoid somata predominated in the cortex surrounding the sulci. Their sparsely ramified axons were unmyelinated and exhibited NPFF-positive bead-like varicosities. We found significantly fewer NPFF-immunopositive cells in the gray matter of the frontal, cingulate, and superior temporal gyri of both sporadic ALS and late-stage AD patients than in controls, and significantly fewer NPFF-positive cells in the subjacent as well as deep white matter of the frontal gyrus of these patients compared to controls. Notably, the number of NPFF-positive cells was also significantly lower in the hippocampal formation in AD compared to controls. In PiD, NPFF-positive cells were present in significantly lower numbers in the gray and white matter of the cingulate and frontal gyrii in comparison to controls. In schizophrenic patients, lower wNPFF cell counts in the neocortex were significant and global (cingulate, frontal, superior temporal gyrus, medial, and inferior gyri). The precise functions of NPFF-positive cells and their relationship to the superficial corticocortical white matter U-fibers are currently unknown. Here, NPFF immunohistochemistry and expression characterize a previously unrecognized population of cells in the human brain, thereby providing a new entry-point for investigating their physiological and pathophysiological roles.
Collapse
Affiliation(s)
- Diana Wiesner
- Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany
- DZNE, Ulm Site, 89081, Ulm, Germany
| | - Simone Feldengut
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany
| | - Sarah Woelfle
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Tobias M Boeckers
- DZNE, Ulm Site, 89081, Ulm, Germany
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | | | - Francesco Roselli
- Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany.
- DZNE, Ulm Site, 89081, Ulm, Germany.
| | - Kelly Del Tredici
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, Ulm University, 89081, Ulm, Germany
| |
Collapse
|
2
|
I F. The unique neuropathological vulnerability of the human brain to aging. Ageing Res Rev 2023; 87:101916. [PMID: 36990284 DOI: 10.1016/j.arr.2023.101916] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD)-related neurofibrillary tangles (NFT), argyrophilic grain disease (AGD), aging-related tau astrogliopathy (ARTAG), limbic predominant TDP-43 proteinopathy (LATE), and amygdala-predominant Lewy body disease (LBD) are proteinopathies that, together with hippocampal sclerosis, progressively appear in the elderly affecting from 50% to 99% of individuals aged 80 years, depending on the disease. These disorders usually converge on the same subject and associate with additive cognitive impairment. Abnormal Tau, TDP-43, and α-synuclein pathologies progress following a pattern consistent with an active cell-to-cell transmission and abnormal protein processing in the host cell. However, cell vulnerability and transmission pathways are specific for each disorder, albeit abnormal proteins may co-localize in particular neurons. All these alterations are unique or highly prevalent in humans. They all affect, at first, the archicortex and paleocortex to extend at later stages to the neocortex and other regions of the telencephalon. These observations show that the phylogenetically oldest areas of the human cerebral cortex and amygdala are not designed to cope with the lifespan of actual humans. New strategies aimed at reducing the functional overload of the human telencephalon, including optimization of dream repair mechanisms and implementation of artificial circuit devices to surrogate specific brain functions, appear promising.
Collapse
Affiliation(s)
- Ferrer I
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain; Biomedical Research Network of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
3
|
Soleimani-Meigooni DN, Iaccarino L, La Joie R, Baker S, Bourakova V, Boxer AL, Edwards L, Eser R, Gorno-Tempini ML, Jagust WJ, Janabi M, Kramer JH, Lesman-Segev OH, Mellinger T, Miller BL, Pham J, Rosen HJ, Spina S, Seeley WW, Strom A, Grinberg LT, Rabinovici GD. 18F-flortaucipir PET to autopsy comparisons in Alzheimer's disease and other neurodegenerative diseases. Brain 2020; 143:3477-3494. [PMID: 33141172 PMCID: PMC7719031 DOI: 10.1093/brain/awaa276] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022] Open
Abstract
Few studies have evaluated the relationship between in vivo18F-flortaucipir PET and post-mortem pathology. We sought to compare antemortem 18F-flortaucipir PET to neuropathology in a consecutive series of patients with a broad spectrum of neurodegenerative conditions. Twenty patients were included [mean age at PET 61 years (range 34-76); eight female; median PET-to-autopsy interval of 30 months (range 4-59 months)]. Eight patients had primary Alzheimer's disease pathology, nine had non-Alzheimer tauopathies (progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, and frontotemporal lobar degeneration with MAPT mutations), and three had non-tau frontotemporal lobar degeneration. Using an inferior cerebellar grey matter reference, 80-100-min 18F-flortaucipir PET standardized uptake value ratio (SUVR) images were created. Mean SUVRs were calculated for progressive supranuclear palsy, corticobasal degeneration, and neurofibrillary tangle Braak stage regions of interest, and these values were compared to SUVRs derived from young, non-autopsy, cognitively normal controls used as a standard for tau negativity. W-score maps were generated to highlight areas of increased tracer retention compared to cognitively normal controls, adjusting for age as a covariate. Autopsies were performed blinded to PET results. There was excellent correspondence between areas of 18F-flortaucipir retention, on both SUVR images and W-score maps, and neurofibrillary tangle distribution in patients with primary Alzheimer's disease neuropathology. Patients with non-Alzheimer tauopathies and non-tau frontotemporal lobar degeneration showed a range of tracer retention that was less than Alzheimer's disease, though higher than age-matched, cognitively normal controls. Overall, binding across both tau-positive and tau-negative non-Alzheimer disorders did not reliably correspond with post-mortem tau pathology. 18F-flortaucipir SUVRs in subcortical regions were higher in autopsy-confirmed progressive supranuclear palsy and corticobasal degeneration than in controls, but were similar to values measured in Alzheimer's disease and tau-negative neurodegenerative pathologies. Quantification of 18F-flortaucipir SUVR images at Braak stage regions of interest reliably detected advanced Alzheimer's (Braak VI) pathology. However, patients with earlier Braak stages (Braak I-IV) did not show elevated tracer uptake in these regions compared to young, tau-negative controls. In summary, PET-to-autopsy comparisons confirm that 18F-flortaucipir PET is a reliable biomarker of advanced Braak tau pathology in Alzheimer's disease. The tracer cannot reliably differentiate non-Alzheimer tauopathies and may not detect early Braak stages of neurofibrillary tangle pathology.
Collapse
Affiliation(s)
- David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Suzanne Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Viktoriya Bourakova
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rana Eser
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | | | - William J Jagust
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Mustafa Janabi
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Orit H Lesman-Segev
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Taylor Mellinger
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Julie Pham
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
4
|
Ferrer I, Andrés-Benito P, Sala-Jarque J, Gil V, Del Rio JA. Capacity for Seeding and Spreading of Argyrophilic Grain Disease in a Wild-Type Murine Model; Comparisons With Primary Age-Related Tauopathy. Front Mol Neurosci 2020; 13:101. [PMID: 32670019 PMCID: PMC7326954 DOI: 10.3389/fnmol.2020.00101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/11/2020] [Indexed: 01/08/2023] Open
Abstract
Argyrophilic grain disease (AGD) is a common 4R-tauopathy, causing or contributing to cognitive impairment in the elderly. AGD is characterized neuropathologically by pre-tangles in neurons, dendritic swellings called grains, threads, thorn-shaped astrocytes, and coiled bodies in oligodendrocytes in the limbic system. AGD has a characteristic pattern progressively involving the entorhinal cortex, amygdala, hippocampus, dentate gyrus, presubiculum, subiculum, hypothalamic nuclei, temporal cortex, and neocortex and brainstem, thus suggesting that argyrophilic grain pathology is a natural model of tau propagation. One series of WT mice was unilaterally inoculated in the hippocampus with sarkosyl-insoluble and sarkosyl-soluble fractions from “pure” AGD at the age of 3 or 7/12 months and killed 3 or 7 months later. Abnormal hyper-phosphorylated tau deposits were found in ipsilateral hippocampal neurons, grains (dots) in the hippocampus, and threads, dots and coiled bodies in the fimbria, as well as the ipsilateral and contralateral corpus callosum. The extension of lesions was wider in animals surviving 7 months compared with those surviving 3 months. Astrocytic inclusions were not observed at any time. Tau deposits were mainly composed of 4Rtau, but also 3Rtau. For comparative purposes, another series of WT mice was inoculated with sarkosyl-insoluble fractions from primary age-related tauopathy (PART), a pure neuronal neurofibrillary tangle 3Rtau + 4Rtau tauopathy involving the deep temporal cortex and limbic system. Abnormal hyper-phosphorylated tau deposits were found in neurons in the ipsilateral hippocampus, coiled bodies and threads in the fimbria, and the ipsilateral and contralateral corpus callosum, which extended with time along the anterior-posterior axis and distant regions such as hypothalamic nuclei and nuclei of the septum when comparing mice surviving 7 months with mice surviving 3 months. Astrocytic inclusions were not observed. Tau deposits were mainly composed of 4Rtau and 3Rtau. These results show the capacity for seeding and spreading of AGD tau and PART tau in the brain of WT mouse, and suggest that characteristics of host tau, in addition to those of inoculated tau, are key to identifying commonalities and differences between human tauopathies and corresponding murine models.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Bellvitge University Hospital, IDIBELL (Bellvitge Biomedical Research Centre), Barcelona, Spain.,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Pol Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Bellvitge University Hospital, IDIBELL (Bellvitge Biomedical Research Centre), Barcelona, Spain.,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Julia Sala-Jarque
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Institute for Science and Technology, Parc Científic de Barcelona, Barcelona, Spain
| | - Vanessa Gil
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - José Antonio Del Rio
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Institute for Science and Technology, Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Jellinger KA. Neuropathology and pathogenesis of extrapyramidal movement disorders: a critical update-I. Hypokinetic-rigid movement disorders. J Neural Transm (Vienna) 2019; 126:933-995. [PMID: 31214855 DOI: 10.1007/s00702-019-02028-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
Abstract
Extrapyramidal movement disorders include hypokinetic rigid and hyperkinetic or mixed forms, most of them originating from dysfunction of the basal ganglia (BG) and their information circuits. The functional anatomy of the BG, the cortico-BG-thalamocortical, and BG-cerebellar circuit connections are briefly reviewed. Pathophysiologic classification of extrapyramidal movement disorder mechanisms distinguish (1) parkinsonian syndromes, (2) chorea and related syndromes, (3) dystonias, (4) myoclonic syndromes, (5) ballism, (6) tics, and (7) tremor syndromes. Recent genetic and molecular-biologic classifications distinguish (1) synucleinopathies (Parkinson's disease, dementia with Lewy bodies, Parkinson's disease-dementia, and multiple system atrophy); (2) tauopathies (progressive supranuclear palsy, corticobasal degeneration, FTLD-17; Guamian Parkinson-dementia; Pick's disease, and others); (3) polyglutamine disorders (Huntington's disease and related disorders); (4) pantothenate kinase-associated neurodegeneration; (5) Wilson's disease; and (6) other hereditary neurodegenerations without hitherto detected genetic or specific markers. The diversity of phenotypes is related to the deposition of pathologic proteins in distinct cell populations, causing neurodegeneration due to genetic and environmental factors, but there is frequent overlap between various disorders. Their etiopathogenesis is still poorly understood, but is suggested to result from an interaction between genetic and environmental factors. Multiple etiologies and noxious factors (protein mishandling, mitochondrial dysfunction, oxidative stress, excitotoxicity, energy failure, and chronic neuroinflammation) are more likely than a single factor. Current clinical consensus criteria have increased the diagnostic accuracy of most neurodegenerative movement disorders, but for their definite diagnosis, histopathological confirmation is required. We present a timely overview of the neuropathology and pathogenesis of the major extrapyramidal movement disorders in two parts, the first one dedicated to hypokinetic-rigid forms and the second to hyperkinetic disorders.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
6
|
Abstract
This chapter describes the visual problems likely to be encountered in Parkinson's disease (PD) and whether such signs are useful in differentiating the parkinsonian syndromes. Visual dysfunction in PD may involve visual acuity, contrast sensitivity, color discrimination, pupil reactivity, saccadic and pursuit eye movements, motion perception, visual fields, and visual processing speeds. In addition, disturbance of visuospatial orientation, facial recognition problems, rapid eye movement (REM) sleep behavior disorder, and chronic visual hallucinations may be present. Problems affecting pupil reactivity, stereopsis, pursuit eye movement, and visuomotor adaptation, when accompanied by REM sleep behavior disorder, could be early features of PD. Dementia associated with PD is associated with enhanced eye movement problems, visuospatial deficits, and visual hallucinations. Visual dysfunction may be a useful diagnostic feature in differentiating PD from other parkinsonian symptoms, visual hallucinations, visuospatial dysfunction, and variation in saccadic eye movement problems being particularly useful discriminating features.
Collapse
|
7
|
Kasahata N, Sato T, Onishi I, Kitagawa M, Uchihara T, Hirokawa K. Three-Repeat Tau with Grain-Like Structures and Distribution in an 83-Year-Old Man. J Alzheimers Dis 2017; 58:681-685. [PMID: 28453470 DOI: 10.3233/jad-160672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We encountered an 83-year-old man with 3-repeat dominant grain-like tau deposition. Tau-positive lesions exhibited apparent similarity to argyrophilic grains in terms of their distribution in the ambient gyrus, amygdala, and dorsomedial temporal tip and the characteristic comma-like morphology. The abundant oligodendroglial tau immunoreactivities were 3-repeat dominant. Tuft-shaped astrocytes showed partial 3-repeat tau immnoreactivities. These grain-like structures, as well as tuft-shaped astrocytes and oligodendroglia, exhibited predominant 3-repeat tau immunoreactivity, suggesting that grain-like structures and their characteristic distribution are mutually linked and not unique to 4-repeat tau deposition. pTDP immunoreactivity, extensive macrophage infiltration, and spongiosis were associated with these 3-repeat tau deposits.
Collapse
Affiliation(s)
- Naoki Kasahata
- Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Medicine, Division of Neurology, Tokyo Metropolitan Ohtsuka Hospital, Tokyo, Japan
| | - Tomohide Sato
- Department of Pulmonary Medicine, Nitobe Memorial Nakano General Hospital, Tokyo, Japan
| | - Iichiroh Onishi
- Department of Pathology, Tokyo Medical Dental University, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Pathology, Tokyo Medical Dental University, Tokyo, Japan
| | - Toshiki Uchihara
- Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Katsuiku Hirokawa
- Department of Pathology, Nitobe Memorial Nakano General Hospital, Tokyo, Japan
| |
Collapse
|
8
|
Armstrong RA, McKee AC, Stein TD, Alvarez VE, Cairns NJ. A quantitative study of tau pathology in 11 cases of chronic traumatic encephalopathy. Neuropathol Appl Neurobiol 2017; 43:154-166. [PMID: 26998921 PMCID: PMC5104666 DOI: 10.1111/nan.12323] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/10/2016] [Accepted: 03/21/2016] [Indexed: 12/14/2022]
Abstract
AIMS To quantify tau pathology of chronic traumatic encephalopathy (CTE) and investigate influence of dot-like lesions (DL), brain region, comorbidity and sporting career length. METHODS Densities of neurofibrillary tangles (NFT), astrocytic tangles (AT), DL, oligodendroglial inclusions (GI), neuropil threads (NT), vacuoles, neurons and enlarged neurons (EN) were measured in tau-immunoreactive sections of upper cortical laminae of frontal and temporal lobes, hippocampus (HC), amygdala and substantia nigra (SN) in 11 cases of CTE. RESULTS DL were a consistent finding in CTE. Densities of NFT, NT and DL were greatest in sectors CA1 and CA2 of the HC. Densities of AT were lower than NFT, small numbers of GI were recorded in temporal lobe and low densities of vacuoles and EN were consistently present. β-Amyloid-containing neuritic plaques (NP) also occurred at low density. Densities of NFT, NT, DL and AT were greater in sulci than gyri, while vacuole density was greater in gyri. Principal components analysis (PCA) suggested that sporting career length and densities of NFT in entorhinal cortex, NT in CA2 and SN and vacuolation in the DG were significant sources of variation among cases. CONCLUSION DL are frequent in CTE suggesting affinity with argyrophilic grain disease (AGD) and Parkinson's disease dementia (PD-Dem). Densities of AT in all regions and NT/DL in sectors CA2/4 were consistent features of CTE. The 11 cases are neuropathologically heterogeneous which may result from genetic diversity, and variation in anatomical pathways subjected to trauma.
Collapse
Affiliation(s)
| | - A C McKee
- VA Boston HealthCare System, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - T D Stein
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
| | - V E Alvarez
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
| | - N J Cairns
- Departments of Neurology and Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
9
|
Kovacs GG, Kwong LK, Grossman M, Irwin DJ, Lee EB, Robinson JL, Suh E, Van Deerlin VM, Lee VM, Trojanowski JQ. Tauopathy with hippocampal 4-repeat tau immunoreactive spherical inclusions: a report of three cases. Brain Pathol 2017; 28:274-283. [PMID: 28019685 DOI: 10.1111/bpa.12482] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/12/2016] [Indexed: 11/29/2022] Open
Abstract
Tauopathies are a major group of neurodegenerative proteinopathies characterized by the accumulation of abnormal and hyperphosphorylated tau proteins in the brain. Tau pathology is characterized as 3R (repeat) or 4R predominant or mixed 3R and 4R type. Here we report three cases lacking mutations in the microtubule associated protein tau (MAPT) gene with unusual tau pathology. The age at onset and duration of illness, respectively, were 63 and 20 years (male), 67 and 5 years (female) and 72 and 20 years (female). The clinical presentation was compatible with a diagnosis of progressive supranuclear palsy (PSP) in two subjects and with cognitive decline in all three subjects. Common neuropathological features included neuronal loss in the hippocampus and dentate gyrus associated with spherical basophilic Pick body-like inclusions showing 4R tau immunoreactivity, which was supported by the detection of predominantly 4R tau species by Western blot examination. In addition, accumulation of tau immunoreactive argyrophilic astrocytes in the hippocampus and amygdala and oligodendroglial coiled bodies in the hippocampal white matter were observed. These morphologies appeared in combination with Alzheimer disease-related pathology and subcortical tau pathology compatible with PSP. Together with a single case report in the literature, our observations on these three cases expand the spectrum of previously described tauopathies. We suggest that this tauopathy variant with hippocampal 4R tau immunoreactive spherical inclusions might contribute to the cognitive deficits in the patients reported here. The precise definition of the clinicopathological relevance of these unusual tau pathologies merits further study.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA.,Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Linda K Kwong
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Murray Grossman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Edward B Lee
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - John L Robinson
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Eunran Suh
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Vivianna M Van Deerlin
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Virginia M Lee
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| |
Collapse
|
10
|
Irwin DJ, Cairns NJ, Grossman M, McMillan CT, Lee EB, Van Deerlin VM, Lee VMY, Trojanowski JQ. Frontotemporal lobar degeneration: defining phenotypic diversity through personalized medicine. Acta Neuropathol 2015; 129:469-91. [PMID: 25549971 PMCID: PMC4369168 DOI: 10.1007/s00401-014-1380-1] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 12/18/2014] [Accepted: 12/20/2014] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) comprises two main classes of neurodegenerative diseases characterized by neuronal/glial proteinaceous inclusions (i.e., proteinopathies) including tauopathies (i.e., FTLD-Tau) and TDP-43 proteinopathies (i.e., FTLD-TDP) while other very rare forms of FTLD are known such as FTLD with FUS pathology (FTLD-FUS). This review focuses mainly on FTLD-Tau and FLTD-TDP, which may present as several clinical syndromes: a behavioral/dysexecutive syndrome (behavioral variant frontotemporal dementia); language disorders (primary progressive aphasia variants); and motor disorders (amyotrophic lateral sclerosis, corticobasal syndrome, progressive supranuclear palsy syndrome). There is considerable heterogeneity in clinical presentations of underlying neuropathology and current clinical criteria do not reliably predict underlying proteinopathies ante-mortem. In contrast, molecular etiologies of hereditary FTLD are consistently associated with specific proteinopathies. These include MAPT mutations with FTLD-Tau and GRN, C9orf72, VCP and TARDBP with FTLD-TDP. The last decade has seen a rapid expansion in our knowledge of the molecular pathologies associated with this clinically and neuropathologically heterogeneous group of FTLD diseases. Moreover, in view of current limitations to reliably diagnose specific FTLD neuropathologies prior to autopsy, we summarize the current state of the science in FTLD biomarker research including neuroimaging, biofluid and genetic analyses. We propose that combining several of these biomarker modalities will improve diagnostic specificity in FTLD through a personalized medicine approach. The goals of these efforts are to enhance power for clinical trials focused on slowing or preventing progression of spread of tau, TDP-43 and other FTLD-associated pathologies and work toward the goal of defining clinical endophenotypes of FTD.
Collapse
Affiliation(s)
- David J Irwin
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Frontotemporal Degeneration Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nigel J. Cairns
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Murray Grossman
- Penn Frontotemporal Degeneration Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Corey T. McMillan
- Penn Frontotemporal Degeneration Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward B. Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vivianna M. Van Deerlin
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M.-Y. Lee
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Q. Trojanowski
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
Agosta F, Galantucci S, Magnani G, Marcone A, Martinelli D, Antonietta Volontè M, Riva N, Iannaccone S, Ferraro PM, Caso F, Chiò A, Comi G, Falini A, Filippi M. MRI signatures of the frontotemporal lobar degeneration continuum. Hum Brain Mapp 2015; 36:2602-14. [PMID: 25821176 DOI: 10.1002/hbm.22794] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 02/16/2015] [Accepted: 03/11/2015] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To identify overlapping and unique grey (GM) and white matter (WM) signatures within the frontotemporal lobar degeneration (FTLD) continuum, and discriminate likely FTLD-TAU and FTLD-TDP patients using structural and diffusion tensor (DT) magnetic resonance imaging (MRI). METHODS T1-weighted and DT MRI were collected from 121 subjects: 35 motor neuron disease (MND), 14 behavioral variant of frontotemporal dementia, 12 semantic and 11 nonfluent primary progressive aphasia, 21 progressive supranuclear palsy syndrome patients, and 28 healthy controls. Patterns of GM atrophy were established using voxel-based morphometry. Tract-based spatial statistics was used to perform a WM voxelwise analysis of mean diffusivity and fractional anisotropy. RESULTS In all clinical FTLD phenotypes, the pattern of WM damage was more distributed than that of GM atrophy. All patient groups, with the exception of MND cases with a pure motor syndrome, shared a focal GM atrophy centered around the dorsolateral and medial frontal cortex and a largely overlapping pattern of WM damage involving the genu and body of the corpus callosum and ventral frontotemporal and dorsal frontoparietal WM pathways. Surrounding this common area, phenotype (symptom)-specific GM and WM regions of damage were found in each group. CONCLUSIONS In the FTLD spectrum, WM disruption is more severe than GM damage. Frontal cortex and WM pathways represent the common target of neurodegeneration in these conditions. The topographic pattern of damage supports a "prion-like" protein propagation through WM connections as underlying mechanism of the stereotyped progression of FTLD.
Collapse
Affiliation(s)
| | | | - Giuseppe Magnani
- Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience
| | - Alessandra Marcone
- Department of Clinical Neurosciences, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | | | | | - Nilo Riva
- Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience
| | - Sandro Iannaccone
- Department of Clinical Neurosciences, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | | | | | - Adriano Chiò
- Department of Neuroscience, ALS Center, "Rita Levi Montalcini" University of Torino, Torino, Italy
| | - Giancarlo Comi
- Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience
| | - Andrea Falini
- Department of Neuroradiology and CERMAC, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience
| |
Collapse
|
12
|
Rockenstein E, Overk CR, Ubhi K, Mante M, Patrick C, Adame A, Bisquert A, Trejo-Morales M, Spencer B, Masliah E. A novel triple repeat mutant tau transgenic model that mimics aspects of pick's disease and fronto-temporal tauopathies. PLoS One 2015; 10:e0121570. [PMID: 25803611 PMCID: PMC4372415 DOI: 10.1371/journal.pone.0121570] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 02/13/2015] [Indexed: 01/18/2023] Open
Abstract
Tauopathies are a group of disorders leading to cognitive and behavioral impairment in the aging population. While four-repeat (4R) Tau is more abundant in corticobasal degeneration, progressive supranuclear palsy, and Alzheimer’s disease, three-repeat (3R) Tau is the most abundant splice, in Pick's disease. A number of transgenic models expressing wild-type and mutant forms of the 4R Tau have been developed. However, few models of three-repeat Tau are available. A transgenic mouse model expressing three-repeat Tau was developed bearing the mutations associated with familial forms of Pick's disease (L266V and G272V mutations). Two lines expressing high (Line 13) and low (Line 2) levels of the three-repeat mutant Tau were analyzed. By Western blot, using antibodies specific to three-repeat Tau, Line 13 expressed 5-times more Tau than Line 2. The Tau expressed by these mice was most abundant in the frontal-temporal cortex and limbic system and was phosphorylated at residues detected by the PHF-1, AT8, CP9 and CP13 antibodies. The higher-expressing mice displayed hyperactivity, memory deficits in the water maze and alterations in the round beam. The behavioral deficits started at 6-8 months of age and were associated with a progressive increase in the accumulation of 3R Tau. By immunocytochemistry, mice from Line 13 displayed extensive accumulation of 3R Tau in neuronal cells bodies in the pyramidal neurons of the neocortex, CA1-3 regions, and dentate gyrus of the hippocampus. Aggregates in the granular cells had a globus appearance and mimic Pick’s-like inclusions. There were abundant dystrophic neurites, astrogliosis and synapto-dendritic damage in the neocortex and hippocampus of the higher expresser line. The hippocampal lesions were moderately argyrophilic and Thioflavin-S negative. By electron microscopy, discrete straight filament aggregates were detected in some neurons in the hippocampus. This model holds promise for better understanding the natural history and progression of 3R tauopathies and their relationship with mitochondrial alterations and might be suitable for therapeutical testing.
Collapse
Affiliation(s)
- Edward Rockenstein
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Cassia R. Overk
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Kiren Ubhi
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Michael Mante
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Christina Patrick
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Anthony Adame
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Alejandro Bisquert
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Margarita Trejo-Morales
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Eliezer Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
13
|
Boluda S, Iba M, Zhang B, Raible KM, Lee VMY, Trojanowski JQ. Differential induction and spread of tau pathology in young PS19 tau transgenic mice following intracerebral injections of pathological tau from Alzheimer's disease or corticobasal degeneration brains. Acta Neuropathol 2015; 129:221-37. [PMID: 25534024 DOI: 10.1007/s00401-014-1373-0] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/04/2014] [Accepted: 12/04/2014] [Indexed: 11/26/2022]
Abstract
Filamentous tau pathologies are hallmark lesions of several neurodegenerative tauopathies including Alzheimer's disease (AD) and corticobasal degeneration (CBD) which show cell type-specific and topographically distinct tau inclusions. Growing evidence supports templated transmission of tauopathies through functionally interconnected neuroanatomical pathways suggesting that different self-propagating strains of pathological tau could account for the diverse manifestations of neurodegenerative tauopathies. Here, we describe the rapid and distinct cell type-specific spread of pathological tau following intracerebral injections of CBD or AD brain extracts enriched in pathological tau (designated CBD-Tau and AD-Tau, respectively) in young human mutant P301S tau transgenic (Tg) mice (line PS19) ~6-9 months before they show onset of mutant tau transgene-induced tau pathology. At 1 month post-injection of CBD-Tau, tau inclusions developed predominantly in oligodendrocytes of the fimbria and white matter near the injection sites with infrequent intraneuronal tau aggregates. In contrast, injections of AD-Tau in young PS19 mice induced tau pathology predominantly in neuronal perikarya with little or no oligodendrocyte involvement 1 month post-injection. With longer post-injection survival intervals of up to 6 months, CBD-Tau- and AD-Tau-induced tau pathology spread to different brain regions distant from the injection sites while maintaining the cell type-specific pattern noted above. Finally, CA3 neuron loss was detected 3 months post-injection of AD-Tau but not CBD-Tau. Thus, AD-Tau and CBD-Tau represent specific pathological tau strains that spread differentially and may underlie distinct clinical and pathological features of these two tauopathies. Hence, these strains could become targets to develop disease-modifying therapies for CBD and AD.
Collapse
Affiliation(s)
- Susana Boluda
- Department of Pathology and Laboratory Medicine, The Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Perelman School of Medicine, 3600 Spruce Street, Philadelphia, PA, 19104-4283, USA
| | | | | | | | | | | |
Collapse
|
14
|
Mari Z, Halls AJ, Vortmeyer A, Zhukareva V, Uryu K, Lee VM, Hallett M. Clinico-Pathological Correlation in Progressive Ataxia and Palatal Tremor: A Novel Tauopathy. Mov Disord Clin Pract 2014; 1:50-56. [PMID: 30363828 PMCID: PMC6183254 DOI: 10.1002/mdc3.12014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/13/2014] [Accepted: 02/16/2014] [Indexed: 12/29/2022] Open
Abstract
Palatal tremor (PT) is an uncommon movement disorder that has been subdivided into essential and symptomatic forms. A distinct subgroup of the symptomatic form presents with progressive ataxia and PT. The histopathology of progressive ataxia and PT has not been previously determined. This study consisted of a clinical review, histopathology, and electron microscopy of the brain of a man with progressive ataxia and PT. The inferior olivary hypertrophy was symmetrical and homogenous, and no focal pathologic lesions could be identified in the brainstem. Insoluble tau deposits were found in neurons, exclusively infratentorially. We present the clinical and pathological evaluation of a case of progressive ataxia and PT that provide evidence for a unique form of 4R tauopathy.
Collapse
Affiliation(s)
- Zoltan Mari
- Human Motor Control SectionMedical Neurology BranchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Andrew J.M. Halls
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Alexander Vortmeyer
- Surgical Neurology BranchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Victoria Zhukareva
- Center for Neurodegenerative Disease ResearchDepartment of Pathology and Laboratory MedicineUniversity of Pennsylvania School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Neurobiology and AnatomyDrexel University School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Kunihiro Uryu
- Center for Neurodegenerative Disease ResearchDepartment of Pathology and Laboratory MedicineUniversity of Pennsylvania School of MedicinePhiladelphiaPennsylvaniaUSA
- Electron Microscopy Resource CenterThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Virginia M.‐Y. Lee
- Center for Neurodegenerative Disease ResearchDepartment of Pathology and Laboratory MedicineUniversity of Pennsylvania School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Mark Hallett
- Human Motor Control SectionMedical Neurology BranchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
15
|
Armstrong RA, Kotzbauer PT, Perlmutter JS, Campbell MC, Hurth KM, Schmidt RE, Cairns NJ. A quantitative study of α-synuclein pathology in fifteen cases of dementia associated with Parkinson disease. J Neural Transm (Vienna) 2014; 121:171-81. [PMID: 23996276 PMCID: PMC4041534 DOI: 10.1007/s00702-013-1084-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 08/19/2013] [Indexed: 12/22/2022]
Abstract
The α-synuclein-immunoreactive pathology of dementia associated with Parkinson disease (DPD) comprises Lewy bodies (LB), Lewy neurites (LN), and Lewy grains (LG). The densities of LB, LN, LG together with vacuoles, neurons, abnormally enlarged neurons (EN), and glial cell nuclei were measured in fifteen cases of DPD. Densities of LN and LG were up to 19 and 70 times those of LB, respectively, depending on region. Densities were significantly greater in amygdala, entorhinal cortex (EC), and sectors CA2/CA3 of the hippocampus, whereas middle frontal gyrus, sector CA1, and dentate gyrus were least affected. Low densities of vacuoles and EN were recorded in most regions. There were differences in the numerical density of neurons between regions, but no statistical difference between patients and controls. In the cortex, the density of LB and vacuoles was similar in upper and lower laminae, while the densities of LN and LG were greater in upper cortex. The densities of LB, LN, and LG were positively correlated. Principal components analysis suggested that DPD cases were heterogeneous with pathology primarily affecting either hippocampus or cortex. The data suggest in DPD: (1) ratio of LN and LG to LB varies between regions, (2) low densities of vacuoles and EN are present in most brain regions, (3) degeneration occurs across cortical laminae, upper laminae being particularly affected, (4) LB, LN and LG may represent degeneration of the same neurons, and (5) disease heterogeneity may result from variation in anatomical pathway affected by cell-to-cell transfer of α-synuclein.
Collapse
|
16
|
Povellato G, Tuxworth RI, Hanger DP, Tear G. Modification of the Drosophila model of in vivo Tau toxicity reveals protective phosphorylation by GSK3β. Biol Open 2014; 3:1-11. [PMID: 24429107 PMCID: PMC3892155 DOI: 10.1242/bio.20136692] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/29/2013] [Indexed: 01/17/2023] Open
Abstract
Hyperphosphorylation of the microtubule associated protein, Tau, is the hallmark of a group of neurodegenerative disorders known as the tauopathies which includes Alzheimer's disease. Precisely how and why Tau phosphorylation is increased in disease is not fully understood, nor how individual sites modify Tau function. Several groups have used the Drosophila visual system as an in vivo model to examine how the toxicity of Tau varies with phosphorylation status. This system relies on overexpression of Tau from transgenes but is susceptible to position effects altering expression and activity of the transgenes. We have refined the system by eliminating position effects through the use of site-specific integration. By standardising Tau expression levels we have been able to compare directly the toxicity of different isoforms of Tau and Tau point mutants that abolish important phosphorylation events. We have also examined the importance of human kinases in modulating Tau toxicity in vivo. We were able to confirm that human GSK3β phosphorylates Tau and increases toxicity but, unexpectedly, we identified that preventing phosphorylation of Ser404 is a protective event. When phosphorylation at this site is prevented, Tau toxicity in the Drosophila visual system is increased in the presence of GSK3β. Our data suggest that not all phosphorylation events on Tau are associated with toxicity.
Collapse
Affiliation(s)
- Giulia Povellato
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, UK
| | - Richard I. Tuxworth
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, UK
- School of Clinical and Experimental Medicine, University of Birmingham, The Medical School, Birmingham B15 2TT, UK
| | - Diane P. Hanger
- Department of Neuroscience, King's College London, Institute of Psychiatry, De Crespigny Park, London SE5 8AF, UK
| | - Guy Tear
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, UK
| |
Collapse
|
17
|
Irwin DJ, Cohen TJ, Grossman M, Arnold SE, McCarty-Wood E, Van Deerlin VM, Lee VMY, Trojanowski JQ. Acetylated tau neuropathology in sporadic and hereditary tauopathies. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:344-51. [PMID: 23885714 DOI: 10.1016/j.ajpath.2013.04.025] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/16/2013] [Accepted: 04/25/2013] [Indexed: 12/12/2022]
Abstract
We have recently shown acetylation of tau at lysine residue 280 (AC-K280) to be a disease-specific modification in Alzheimer disease (AD), corticobasal degeneration, and progressive supranuclear palsy, likely representing a major regulatory tau modification. Herein, we extend our observations using IHC with a polyclonal antibody specific for AC-K280. Thirty brain regions were examined in argyrophilic grain disease (AGD; n = 5), tangle-predominant senile dementia (TPSD; n = 5), Pick disease (n = 4), familial AD (FAD; n = 2; PSEN1 p.G206A and p.S170P), and frontotemporal dementia with parkinsonism linked to chromosome-17 (FTDP-17; n = 2; MAPT p.P301L and IVS10 + 16). All AGD, TPSD, FAD, and FTDP-17 cases had significant AC-K280 reactivity that was similar in severity and distribution to phosphorylated tau. AC-K280 robustly labeled grain pathological characteristics in AGD and was predominantly associated with thioflavin-S-positive neurofibrillary tangles and less reactive in neuropil threads and extracellular tangles in TPSD and FAD. Thioflavin-S-negative neuronal and glial inclusions of patients with FTDP-17 were robustly AC-K280 reactive. A low degree of AC-K280 was found in a subset of 4-repeat tau-containing lesions in Pick disease. AC-K280 is a prominent feature of both neuronal and glial tau aggregations in tauopathies of various etiologies. The close association of AC-K280 with amyloid and pre-amyloid conformations of tau suggests a potential role in tangle maturation and, thus, could serve as a useful biomarker or therapeutic target in a variety of tauopathies.
Collapse
Affiliation(s)
- David J Irwin
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-4283, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Rábano A, Cuadros R, Calero M, Hernández F, Avila J. Specific profile of tau isoforms in argyrophylic grain disease. J Exp Neurosci 2013; 7:51-9. [PMID: 25157208 PMCID: PMC4089774 DOI: 10.4137/jen.s12202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Argyrophylic grain disease (AGD) is a neurodegenerative condition that has been classified among the sporadic tauopathies. Entities in this group present intracellular aggregates of hyperphosphorylated tau, giving rise to characteristic neuronal and glial inclusions. In different tauopathies, the proportion of several tau isoforms present in the aggregates shows specific patterns. AGD has been tentatively classified in the 4R group (predominance of 4R tau isoforms) together with progressive supranuclear palsy and corticobasal degeneration. Pick's disease is included in the 3R group (predominance of 3R isoforms), whereas tau pathology of Alzheimer's disease represents and intermediate group (3 or 4 repeats [3R plus 4R, respectively] isoforms). In this work, we have analyzed tau present in aggregates isolated from brain samples of patients with argyrophylic grain disease. Our results indicate that the main tau isoform present in aggregates obtained from patients with AGD is a hyperphosphorylated isoform containing exons 2 and 10 but lacking exon 3.
Collapse
Affiliation(s)
- Alberto Rábano
- Banco de Tejidos de la Fundación CIEN, CIEN Foundation, Carlos III Institute of Health, Alzheimer Center Reina Sofia Foundation, Madrid, Spain
| | - Raquel Cuadros
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Miguel Calero
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Unidad de Encefalopatías Espongiformes, Centro Nacional de Microbiología, Instituto de Salud Carlos III (CNM-ISCIII), Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús Avila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
19
|
Ferrer-Acosta Y, Rodríguez-Cruz EN, Orange F, De Jesús-Cortés H, Madera B, Vaquer-Alicea J, Ballester J, Guinel MJF, Bloom GS, Vega IE. EFhd2 is a novel amyloid protein associated with pathological tau in Alzheimer's disease. J Neurochem 2013; 125:921-31. [PMID: 23331044 PMCID: PMC3676478 DOI: 10.1111/jnc.12155] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 01/08/2013] [Accepted: 01/14/2013] [Indexed: 11/28/2022]
Abstract
EFhd2 is a conserved calcium-binding protein, abundant within the central nervous system. Previous studies identified EFhd2 associated with pathological forms of tau proteins in the tauopathy mouse model JNPL3, which expresses the human tau(P301L) mutant. This association was validated in human tauopathies, such as Alzheimer's disease (AD). However, the role that EFhd2 may play in tauopathies is still unknown. Here, we show that EFhd2 formed amyloid structures in vitro, a capability that is reduced by calcium ions. Electron microscopy (EM) analyses demonstrated that recombinant EFhd2 formed filamentous structures. EM analyses of sarkosyl-insoluble fractions derived from human AD brains also indicated that EFhd2 co-localizes with aggregated tau proteins and formed granular structures. Immunohistological analyses of brain slices demonstrated that EFhd2 co-localizes with pathological tau proteins in AD brains, confirming the co-aggregation of EFhd2 and pathological tau. Furthermore, EFhd2's coiled-coil domain mediated its self-oligomerization in vitro and its association with tau proteins in JNPL3 mouse brain extracts. The results demonstrate that EFhd2 is a novel amyloid protein associated with pathological tau proteins in AD brain and that calcium binding may regulate the formation of EFhd2's amyloid structures. Hence, EFhd2 may play an important role in the pathobiology of tau-mediated neurodegeneration.
Collapse
Affiliation(s)
- Yancy Ferrer-Acosta
- Department of Biology, College of Natural Sciences, University of Puerto Rico, PO Box 23360, San Juan, Puerto Rico 00931-3360, USA
| | - Eva N. Rodríguez-Cruz
- Department of Biology, College of Natural Sciences, University of Puerto Rico, PO Box 23360, San Juan, Puerto Rico 00931-3360, USA
| | - François Orange
- Nanoscopy Facility, College of Natural Sciences, University of Puerto Rico, PO Box 70377, San Juan, Puerto Rico 00936-8377, USA
- Department of Physics, College of Natural Sciences, University of Puerto Rico, PO Box 70377, San Juan, Puerto Rico 00936-8377, USA
| | - Hector De Jesús-Cortés
- Department of Biology, College of Natural Sciences, University of Puerto Rico, PO Box 23360, San Juan, Puerto Rico 00931-3360, USA
| | - Bismark Madera
- Department of Biology, College of Natural Sciences, University of Puerto Rico, PO Box 23360, San Juan, Puerto Rico 00931-3360, USA
- Confocal Microscopy Facility, College of Natural Sciences, University of Puerto Rico – Río Piedras Campus, San Juan, PR 00931
| | - Jaime Vaquer-Alicea
- Department of Biology, College of Natural Sciences, University of Puerto Rico, PO Box 23360, San Juan, Puerto Rico 00931-3360, USA
| | - Juan Ballester
- Department of Biology, College of Natural Sciences, University of Puerto Rico, PO Box 23360, San Juan, Puerto Rico 00931-3360, USA
| | - Maxime J-F. Guinel
- Nanoscopy Facility, College of Natural Sciences, University of Puerto Rico, PO Box 70377, San Juan, Puerto Rico 00936-8377, USA
- Department of Physics, College of Natural Sciences, University of Puerto Rico, PO Box 70377, San Juan, Puerto Rico 00936-8377, USA
- Department of Chemistry, College of Natural Sciences, University of Puerto Rico, PO Box 70377, San Juan, Puerto Rico 00936-8377, USA
| | - George S. Bloom
- Department of Biology, University of Virgina, Charlottesville, VA 22904, USA
| | - Irving E. Vega
- Department of Biology, College of Natural Sciences, University of Puerto Rico, PO Box 23360, San Juan, Puerto Rico 00931-3360, USA
| |
Collapse
|
20
|
Hundelt M, Fath T, Selle K, Oesterwind K, Jordan J, Schultz C, Götz J, von Engelhardt J, Monyer H, Lewejohann L, Sachser N, Bakota L, Brandt R. Altered phosphorylation but no neurodegeneration in a mouse model of tau hyperphosphorylation. Neurobiol Aging 2011; 32:991-1006. [DOI: 10.1016/j.neurobiolaging.2009.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 06/10/2009] [Accepted: 06/27/2009] [Indexed: 01/16/2023]
|
21
|
Abstract
The presence of argyrophilic grains in the neuropil is associated with a form of dementia. We investigated morphological asymmetry in 653 consecutive autopsy patients from a general geriatric hospital (age [mean +/- SD] = 81.1 +/- 8.9 years), focusing on those from patients with advanced argyrophilic grain disease. Paraffin sections of the bilateral posterior hippocampi were immunostained with anti-phosphorylated tau and anti-4-repeat tau antibodies and by the Gallyas-Braak method. In a side-to-side comparison, asymmetry was defined when either the extent or the density of argyrophilic grains was different. Of the 653 subjects, 65 (10%) had Stage 3 argyrophilic grain disease, and 59 (90.8%) showed histopathological asymmetry. Antemortem computed tomographic images (n = 24), magnetic resonance imaging scans (n = 8), and combined computed tomographic and magnetic resonance images (n = 15) were available; images from 20 of the 47 subjects showed asymmetry that correlated with the histopathological asymmetry. Cerebral cortical asymmetry consistent with the histopathology was also visible in N-isopropyl-123I-p-iodoamphetamine single photon emission computed tomographic images from 6 patients and 18F-labeled fluorodeoxyglucose positron emission tomographic images from 2 patients. Thus, asymmetric involvement of the medial temporal lobe in patients with advanced argyrophilic grain disease may represent a diagnostic feature and contribute to distinguishing dementia with grains from Alzheimer disease.
Collapse
|
22
|
Garbern JY, Neumann M, Trojanowski JQ, Lee VMY, Feldman G, Norris JW, Friez MJ, Schwartz CE, Stevenson R, Sima AAF. A mutation affecting the sodium/proton exchanger, SLC9A6, causes mental retardation with tau deposition. ACTA ACUST UNITED AC 2010; 133:1391-402. [PMID: 20395263 DOI: 10.1093/brain/awq071] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We have studied a family with severe mental retardation characterized by the virtual absence of speech, autism spectrum disorder, epilepsy, late-onset ataxia, weakness and dystonia. Post-mortem examination of two males revealed widespread neuronal loss, with the most striking finding being neuronal and glial tau deposition in a pattern reminiscent of corticobasal degeneration. Electron microscopic examination of isolated tau filaments demonstrated paired helical filaments and ribbon-like structures. Biochemical studies of tau demonstrated a preponderance of 4R tau isoforms. The phenotype was linked to Xq26.3, and further analysis identified an in-frame 9 base pair deletion in the solute carrier family 9, isoform A6 (SLC9A6 gene), which encodes sodium/hydrogen exchanger-6 localized to endosomal vesicles. Sodium/hydrogen exchanger-6 is thought to participate in the targeting of intracellular vesicles and may be involved in recycling synaptic vesicles. The striking tau deposition in our subjects reveals a probable interaction between sodium/proton exchangers and cytoskeletal elements involved in vesicular transport, and raises the possibility that abnormalities of vesicular targeting may play an important role in more common disorders such as Alzheimer's disease and autism spectrum disorders.
Collapse
Affiliation(s)
- James Y Garbern
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Absence of alpha-synuclein pathology in postencephalitic parkinsonism. Acta Neuropathol 2009; 118:371-9. [PMID: 19404653 DOI: 10.1007/s00401-009-0537-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 04/09/2009] [Accepted: 04/10/2009] [Indexed: 01/07/2023]
Abstract
Postencephalitic parkinsonism (PEP), a chronic complication of encephalitis lethargica, is a tauopathy characterized by multisystem neuronal loss and gliosis with widespread neurofibrillary lesions composed of both 3- and 4-repeat (3R and 4R) tau isoforms. Previous immunohistochemical studies in a small number of PEP cases demonstrated absence of Lewy bodies as well as the lack of other alpha-synuclein pathology, classifying PEP as a "pure" tauopathy. Neuropathologic examination of 10 brains with clinico-pathologically verified PEP confirmed widespread neurodegeneration in subcortical and brainstem areas associated with multifocal neurofibrillary pathology comprising both 3R and 4R tau. Very rare beta-amyloid deposits were observed in two elderly patients, while Lewy bodies and neurites or any other alpha-synuclein deposits were completely absent. The causes and molecular background of total absence of alpha-synuclein pathology in PEP, in contrast to most other tauopathies, remain as unknown as the pathogenesis of PEP.
Collapse
|
24
|
Neumann M, Rademakers R, Roeber S, Baker M, Kretzschmar HA, Mackenzie IRA. A new subtype of frontotemporal lobar degeneration with FUS pathology. Brain 2009; 132:2922-31. [PMID: 19674978 DOI: 10.1093/brain/awp214] [Citation(s) in RCA: 542] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Frontotemporal dementia (FTD) is a clinical syndrome with a heterogeneous molecular basis. The neuropathology associated with most FTD is characterized by abnormal cellular aggregates of either transactive response DNA-binding protein with Mr 43 kDa (TDP-43) or tau protein. However, we recently described a subgroup of FTD patients, representing around 10%, with an unusual clinical phenotype and pathology characterized by frontotemporal lobar degeneration with neuronal inclusions composed of an unidentified ubiquitinated protein (atypical FTLD-U; aFTLD-U). All cases were sporadic and had early-onset FTD with severe progressive behavioural and personality changes in the absence of aphasia or significant motor features. Mutations in the fused in sarcoma (FUS) gene have recently been identified as a cause of familial amyotrophic lateral sclerosis, with these cases reported to have abnormal cellular accumulations of FUS protein. Because of the recognized clinical, genetic and pathological overlap between FTD and amyotrophic lateral sclerosis, we investigated whether FUS might also be the pathological protein in aFTLD-U. In all our aFTLD-U cases (n = 15), FUS immunohistochemistry labelled all the neuronal inclusions and also demonstrated previously unrecognized glial pathology. Immunoblot analysis of protein extracted from post-mortem aFTLD-U brain tissue demonstrated increased levels of insoluble FUS. No mutations in the FUS gene were identified in any of our patients. These findings suggest that FUS is the pathological protein in a significant subgroup of sporadic FTD and reinforce the concept that FTD and amyotrophic lateral sclerosis are closely related conditions.
Collapse
Affiliation(s)
- Manuela Neumann
- Institute of Neuropathology, University Hospital of Zürich, Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Frontotemporal dementia (FTD) is a clinical syndrome with a heterogeneous molecular basis. Familial FTD has been linked to mutations in several genes, including those encoding the microtubule-associated protein tau (MAPT), progranulin (GRN), valosin-containing protein (VCP) and charged multivescicular body protein 2B (CHMP2B). The associated neuropathology is characterised by selective degeneration of the frontal and temporal lobes (frontotemporal lobar degeneration, FTLD), usually with the presence of abnormal intracellular protein accumulations. The current classification of FTLD neuropathology is based on the identity of the predominant protein abnormality, in the belief that this most closely reflects the underlying pathogenic process. Major subgroups include those characterised by the pathological tau, TDP-43, intermediate filaments and a group with cellular inclusions composed of an unidentified ubiquitinated protein. This review will focus on the current understanding of the molecular basis of each of the major FTLD subtypes. It is anticipated that this knowledge will provide the basis of future advances in the diagnosis and treatment of FTD.
Collapse
|
26
|
Anderson JM, Patani R, Reynolds R, Nicholas R, Compston A, Spillantini MG, Chandran S. Evidence for abnormal tau phosphorylation in early aggressive multiple sclerosis. Acta Neuropathol 2009; 117:583-9. [PMID: 19288121 DOI: 10.1007/s00401-009-0515-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 03/04/2009] [Accepted: 03/05/2009] [Indexed: 11/25/2022]
Abstract
Although progression in multiple sclerosis is pathologically dominated by neurodegeneration, the underlying mechanism is unknown. Abnormal hyperphosphorylation of tau is implicated in the aetiopathogenesis of some common neurodegenerative disorders. We recently demonstrated the association of insoluble tau with established secondary progressive MS, raising the hypothesis that its accumulation is relevant to disease progression. In order to begin to determine the temporal emergence of abnormal tau with disease progression in MS, we examined tau phosphorylation in cerebral tissue from a rare case of early aggressive MS. We report tau hyperphosphorylation occurring in multiple cell types, with biochemical analysis confirming restriction to the soluble fraction. The absence of sarcosyl-insoluble tau fraction in early disease and its presence in secondary progression raises the possibility that insoluble tau accumulates with disease progression.
Collapse
Affiliation(s)
- Jane Marian Anderson
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
27
|
Salat DH, Greve DN, Pacheco JL, Quinn BT, Helmer KG, Buckner RL, Fischl B. Regional white matter volume differences in nondemented aging and Alzheimer's disease. Neuroimage 2008; 44:1247-58. [PMID: 19027860 DOI: 10.1016/j.neuroimage.2008.10.030] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 10/15/2008] [Accepted: 10/16/2008] [Indexed: 11/15/2022] Open
Abstract
Accumulating evidence suggests that altered cerebral white matter (WM) influences normal aging, and further that WM degeneration may modulate the clinical expression of Alzheimer's disease (AD). Here we conducted a study of differences in WM volume across the adult age span and in AD employing a newly developed, automated method for regional parcellation of the subcortical WM that uses curvature landmarks and gray matter (GM)/WM surface boundary information. This procedure measures the volume of gyral WM, utilizing a distance constraint to limit the measurements from extending into the centrum semiovale. Regional estimates were first established to be reliable across two scan sessions in 20 young healthy individuals. Next, the method was applied to a large clinically-characterized sample of 299 individuals including 73 normal older adults and 91 age-matched participants with very mild to mild AD. The majority of measured regions showed a decline in volume with increasing age, with strong effects found in bilateral fusiform, lateral orbitofrontal, superior frontal, medial orbital frontal, inferior temporal, and middle temporal WM. The association between WM volume and age was quadratic in many regions suggesting that WM volume loss accelerates in advanced aging. A number of WM regions were further reduced in AD with parahippocampal, entorhinal, inferior parietal and rostral middle frontal WM showing the strongest AD-associated reductions. There were minimal sex effects after correction for intracranial volume, and there were associations between ventricular volume and regional WM volumes in the older adults and AD that were not apparent in the younger adults. Certain results, such as the loss of WM in the fusiform region with aging, were unexpected and provide novel insight into patterns of age associated neural and cognitive decline. Overall, these results demonstrate the utility of automated regional WM measures in revealing the distinct patterns of age and AD associated volume loss that may contribute to cognitive decline.
Collapse
Affiliation(s)
- David H Salat
- Department of Radiology, Massachusetts General Hospital, Boston MA, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Anderson JM, Hampton DW, Patani R, Pryce G, Crowther RA, Reynolds R, Franklin RJM, Giovannoni G, Compston DAS, Baker D, Spillantini MG, Chandran S. Abnormally phosphorylated tau is associated with neuronal and axonal loss in experimental autoimmune encephalomyelitis and multiple sclerosis. ACTA ACUST UNITED AC 2008; 131:1736-48. [PMID: 18567922 DOI: 10.1093/brain/awn119] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The pathological correlate of clinical disability and progression in multiple sclerosis is neuronal and axonal loss; however, the underlying mechanisms are unknown. Abnormal phosphorylation of tau is a common feature of some neurodegenerative disorders, such as Alzheimer's disease. We investigated the presence of tau hyperphosphorylation and its relationship with neuronal and axonal loss in chronic experimental autoimmune encephalomyelitis (CEAE) and in brain samples from patients with secondary progressive multiple sclerosis. We report the novel finding of abnormal tau phosphorylation in CEAE. We further show that accumulation of insoluble tau is associated with both neuronal and axonal loss that correlates with progression from relapsing-remitting to chronic stages of EAE. Significantly, analysis of secondary progressive multiple sclerosis brain tissue also revealed abnormally phosphorylated tau and the formation of insoluble tau. Together, these observations provide the first evidence implicating abnormal tau in the neurodegenerative phase of tissue injury in experimental and human demyelinating disease.
Collapse
Affiliation(s)
- J M Anderson
- Department of Clinical Neurosciences, Cambridge Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
|
31
|
Tau kinase inhibitors protect hippocampal synapses despite of insoluble tau accumulation. Mol Cell Neurosci 2007; 37:559-67. [PMID: 18201898 DOI: 10.1016/j.mcn.2007.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 11/14/2007] [Accepted: 12/06/2007] [Indexed: 12/25/2022] Open
Abstract
A better understanding of the cellular and molecular pathomechanisms of Alzheimer's disease (AD) is a prerequisite for the development of efficient treatments. We have used a novel assay system based on virus-transduced organotypic hippocampal slice cultures that mimics important aspects of tau-driven AD pathology in a short time frame. Human tau P301L, when expressed in pyramidal neurons of hippocampal slice cultures, was increasingly phosphorylated at several disease-relevant epitopes, leading to progressive neuronal dystrophy and formation of RIPA-insoluble tau. AD-like tau hyperphosphorylation was reduced by the tau kinase inhibitors lithium and SRN-003-556, but RIPA-insoluble tau remained unaffected after treatment with any of these substances. Only SRN-003-556 was able to protect hippocampal neurons from synaptic damage that was presumably caused by a toxic soluble tau fraction. These data provide first mechanistic insights towards the functional benefits of SRN-003-556 that have been observed in vivo.
Collapse
|
32
|
Albasanz JL, Rodríguez A, Ferrer I, Martín M. Up-regulation of adenosine A1 receptors in frontal cortex from Pick's disease cases. Eur J Neurosci 2007; 26:3501-8. [DOI: 10.1111/j.1460-9568.2007.05965.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Mondragón-Rodríguez S, Mena R, Binder LI, Smith MA, Perry G, García-Sierra F. Conformational changes and cleavage of tau in Pick bodies parallel the early processing of tau found in Alzheimer pathology. Neuropathol Appl Neurobiol 2007; 34:62-75. [PMID: 17971079 DOI: 10.1111/j.1365-2990.2007.00853.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neuronal protein inclusions are a common feature in Alzheimer disease (AD) and Pick disease. Even though the inclusions are morphologically different, flame-shape structure for AD vs. spherical structure for Pick disease, both have filaments mainly composed of tau protein. In AD, a well-defined pattern of conformational changes and truncation has been described. In this study, we used laser scanning confocal microscopy to characterize and compare the processing of tau protein during Pick disease with that found in AD. We found that tau protein of Pick disease preserves most of the relevant epitopes found in AD, the conformational foldings labelled by Alz-50 and Tau-66, the cleavage sites D(421) and E(391), as well as many phosphorylated sites, such as Ser(199/202), Thr(205) and Ser(396/404). We found a strong pattern of association between phosphorylation and cleavage at site D(421), as well as the phosphorylation and the conformational Alz-50 epitope. When we used late AD markers such as the conformational Tau-66 epitope and MN423 (cleavage at site E(391)) in Pick bodies (PBs), the overlap was significantly less. Furthermore, following morphological quantification, we found significantly higher numbers of phosphorylated tau in PBs. Overall, our findings suggest that phosphorylation is an early event, likely preceding the cleavage of tau at D(421). Despite this consistency with AD, we found a major distinction, namely that PBs lack beta-sheet conformation. We propose a scheme of early tau processing in these structures, similar to neurofibrillary tangles of AD.
Collapse
|
34
|
Perez-Buira S, Barrachina M, Rodriguez A, Albasanz JL, Martín M, Ferrer I. Expression levels of adenosine receptors in hippocampus and frontal cortex in argyrophilic grain disease. Neurosci Lett 2007; 423:194-9. [PMID: 17707587 DOI: 10.1016/j.neulet.2007.06.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 06/24/2007] [Accepted: 06/25/2007] [Indexed: 12/24/2022]
Abstract
Expression of adenosine receptors of the A1, A2A and A2B type has been examined in the post-mortem frontal cortex and hippocampus in argyrophilic grain disease (AGD), a tauopathy affecting the hippocampus but usually not the frontal cortex, in an attempt to learn about the modulation of the adenosine pathway in this disorder. Significant increased levels of A1, but not of A2A and A2B, have been observed in AGD in the hippocampus but not in the frontal cortex, when compared with age-matched controls. This is accompanied by increased levels of adenylyl cyclase (AC), an effector of A1, and by increased (although not significant) percentage of inhibition of forskolin-stimulated AC by the A1 agonist cyclohexyladenosine in the hippocampus in AGD. These findings indicate sensitization of A1/AC in the hippocampus in AGD, and support a putative activation of the A1/AC pathway that may facilitate protection of this preferentially involved region in AGD.
Collapse
Affiliation(s)
- S Perez-Buira
- Institut de Neuropatologia, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Facultad de Medicina, Universitat de Barcelona, carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | | | | | | | | | | |
Collapse
|
35
|
Jellinger KA, Attems J. Neurofibrillary tangle-predominant dementia: comparison with classical Alzheimer disease. Acta Neuropathol 2007; 113:107-17. [PMID: 17089134 DOI: 10.1007/s00401-006-0156-7] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 10/04/2006] [Accepted: 10/04/2006] [Indexed: 01/23/2023]
Abstract
Neurofibrillary tangle predominant dementia (NFTPD) is a subset of late onset dementia, clinically different from traditional "plaque and tangle" Alzheimer disease (AD): later onset, shorter duration, less severe cognitive impairment, and almost absence of ApoE epsilon4. Neuropathology reveals abundant allocortical neurofibrillary pathology with no or few isocortical tau lesions, absence of neuritic plaques, absence or scarcity of amyloid deposits, but neurofibrillary changes comprising both 3 and 4 repeat (3R and 4R) tau immunohistochemistry are not significantly different from those in classical AD. Comparing 51 autopsy cases of NFTPD with 244 classical AD subjects, the nosology of NFTPD and its differences from AD are discussed.
Collapse
Affiliation(s)
- K A Jellinger
- Institute of Clinical Neurobiology, 18, Kenyongasse, 1070, Vienna, Austria.
| | | |
Collapse
|
36
|
Albasanz JL, Rodríguez A, Ferrer I, Martín M. Adenosine A2A receptors are up-regulated in Pick's disease frontal cortex. Brain Pathol 2007; 16:249-55. [PMID: 17107593 PMCID: PMC8095809 DOI: 10.1111/j.1750-3639.2006.00026.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adenosine A2A receptors (A2AR) are highly expressed in striatum. However, they are also present in extrastriatal structures. A2AR were studied in post-mortem human frontal cortex from Pick's disease (PiD) and age-matched non-demented controls by radioligand binding assays, Western-blotting, real-time PCR and adenylyl cyclase activity determination. Saturation binding assay using [3H]ZM 241385, a selective A2A antagonist, as radioligand revealed a significant increase in total adenosine A2AR numbers (Bmax) in frontal cortex from PiD samples (191% of control Bmax), suggesting up-regulation of this receptor. A significant increase in the level of A2AR was also detected by Western-blotting. Furthermore, expression of mRNA coding A2AR determined by quantitative real-time PCR was enhanced. In agreement, stimulation of adenylyl cyclase by CGS 21680, a selective A2A receptor agonist, was significantly strengthened. Up-regulation of A2B receptors and their corresponding mRNA was also observed. These results show that A2A adenosine receptor/adenylyl cyclase transduction pathway is up-regulated and sensitized in frontal cortex brain from PiD.
Collapse
Affiliation(s)
- José Luís Albasanz
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Químicas, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla‐La Mancha, Ciudad Real, Spain
| | - Agustín Rodríguez
- Departamento de Biología Celular y Anatomía Patológica, Facultad de Medicina, Universidad de Barcelona, campus de Bellvitge, and
| | - Isidro Ferrer
- Departamento de Biología Celular y Anatomía Patológica, Facultad de Medicina, Universidad de Barcelona, campus de Bellvitge, and
- Instituto de Neuropatología, Servicio de Anatomía Patológica, IDIBELL‐Hospital Universitario de Bellvitge, Hospitalet de Llobregat, Spain
| | - Mairena Martín
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Químicas, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla‐La Mancha, Ciudad Real, Spain
| |
Collapse
|
37
|
Zhukareva V, Joyce S, Schuck T, Van Deerlin V, Hurtig H, Albin R, Gilman S, Chin S, Miller B, Trojanowski JQ, Lee VMY. Unexpected abundance of pathological tau in progressive supranuclear palsy white matter. Ann Neurol 2006; 60:335-45. [PMID: 16823854 DOI: 10.1002/ana.20916] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate whether biochemical insoluble tau with 4 (4R) and/or 3 (3R) microtubule-binding repeats accumulate in white as well as gray matter in progressive supranuclear palsy (PSP), a neurodegenerative tauopathy. METHODS To assess tau pathology in PSP white matter, we combined Western blot (WB) and immunohistochemical methods to analyze 23 autopsy-confirmed PSP brains. RESULTS WBs showed an unexpected abundance of insoluble tau in white and gray matter of PSP brains, but biochemical tau pathology in white matter was not correlated with immunohistochemistry using the same panel of epitope-specific anti-tau antibodies used for WB. Despite heterogeneity in the representation of pathological 3R and 4R tau isoforms in cortical versus subcortical regions, biochemically detectable white matter tau pathology is a constant feature of PSP. INTERPRETATION These studies show additional similarities between PSP and corticobasal degeneration, but unlike corticobasal degeneration, more abundant white matter tau pathology in PSP is detectable by WB than by immunohistochemistry. The differential detection of abnormal tau by biochemistry versus microscopy in PSP may reflect distinct pathological mechanisms, and elucidation of these processes will augment efforts to develop better strategies for the diagnosis and treatment of PSP and related neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Victoria Zhukareva
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4283, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Development and implementation of microarray techniques to quantify expression levels of dozens to hundreds to thousands of transcripts simultaneously within select tissue samples from normal control subjects and neurodegenerative diseased brains has enabled scientists to create molecular fingerprints of vulnerable neuronal populations in Alzheimer's disease (AD) and related disorders. A goal is to sample gene expression from homogeneous cell types within a defined region without potential contamination by expression profiles of adjacent neuronal subpopulations and nonneuronal cells. The precise resolution afforded by single cell and population cell RNA analysis in combination with microarrays and real-time quantitative polymerase chain reaction (qPCR)-based analyses allows for relative gene expression level comparisons across cell types under different experimental conditions and disease progression. The ability to analyze single cells is an important distinction from global and regional assessments of mRNA expression and can be applied to optimally prepared tissues from animal models of neurodegeneration as well as postmortem human brain tissues. Gene expression analysis in postmortem AD brain regions including the hippocampal formation and neocortex reveals selectively vulnerable cell types share putative pathogenetic alterations in common classes of transcripts, for example, markers of glutamatergic neurotransmission, synaptic-related markers, protein phosphatases and kinases, and neurotrophins/neurotrophin receptors. Expression profiles of vulnerable regions and neurons may reveal important clues toward the understanding of the molecular pathogenesis of various neurological diseases and aid in identifying rational targets toward pharmacotherapeutic interventions for progressive, late-onset neurodegenerative disorders such as mild cognitive impairment (MCI) and AD.
Collapse
Affiliation(s)
- Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA.
| | | | | | | |
Collapse
|
39
|
Jicha GA, Petersen RC, Knopman DS, Boeve BF, Smith GE, Geda YE, Johnson KA, Cha R, Delucia MW, Braak H, Dickson DW, Parisi JE. Argyrophilic grain disease in demented subjects presenting initially with amnestic mild cognitive impairment. J Neuropathol Exp Neurol 2006; 65:602-9. [PMID: 16783170 DOI: 10.1097/01.jnen.0000225312.11858.57] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
A previous autopsy study of patients with amnestic-type mild cognitive impairment (MCI) suggested an overrepresentation of argyrophilic grain disease (AGD). We studied 34 patients who had diagnoses of amnestic MCI during progression to dementia and who came to autopsy. Neuropathologic evaluation included routine histochemical and immunohistochemical methods, including a 4-repeat tau-specific marker (ET3). AGD was found in association with a variety of neuropathologic diseases in 18 (53%) cases but was the primary pathologic finding in only one (3%) case. ET3 allowed the detection of AGD in 5 additional cases missed using standard techniques. Cases with AGD were significantly older than those without (mean, 94 vs 84 years; p < 0.004, rank sum test). No significant differences were found between groups for other demographic variables, association of AGD with neuropathologic findings of Alzheimer disease, Lewy body, or cerebrovascular disease, or global measures of cognitive function, although there was a nonsignificant trend towards worsening cognitive status in cases with AGD. AGD is a common pathologic finding in subjects who have been diagnosed with amnestic MCI.
Collapse
Affiliation(s)
- Gregory A Jicha
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chau KWK, Chan WY, Shaw PC, Chan HYE. Biochemical investigation of Tau protein phosphorylation status and its solubility properties in Drosophila. Biochem Biophys Res Commun 2006; 346:150-9. [PMID: 16759647 DOI: 10.1016/j.bbrc.2006.05.112] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Accepted: 05/13/2006] [Indexed: 12/20/2022]
Abstract
Tau hyperphosphorylation and insoluble aggregate formation are two cellular features of tauopathies. However, the contribution of Tau protein hyperphosphorylation and its aggregation to Tau pathology still remain controversial. Overexpression of human tau transgenes in the Drosophila eye is toxic and causes neuronal degeneration. We showed that human Tau protein was phosphorylated by endogenous protein kinases in flies, and overexpression of either GSK3beta or Cdk5 enhanced tau-induced toxicity. Using a dominant-negative approach, we showed that kinase activity is important for the enhancement of tau-induced toxicity. Interestingly, such enhancement was accompanied with hyperphosphorylation and alteration of protein solubility properties of Tau. This situation was reminiscent of that observed in pre-tangle neurons in tauopathies patients. We also observed age-dependent Tau aggregate formation in aged transgenic flies. In summary, tau-induced toxicity is enhanced when the human Tau protein undergoes hyperphosphorylation, and we further demonstrated that aging contributes to Tau aggregate formation. Our data also underscore the utilization of transgenic Drosophila Tau models for the studies of pre-tangle events in tauopathies.
Collapse
Affiliation(s)
- Katy Wing-Kam Chau
- Department of Biochemistry and Molecular Biotechnology Programme, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | |
Collapse
|
41
|
Ginsberg SD, Che S, Counts SE, Mufson EJ. Shift in the ratio of three-repeat tau and four-repeat tau mRNAs in individual cholinergic basal forebrain neurons in mild cognitive impairment and Alzheimer's disease. J Neurochem 2006; 96:1401-8. [PMID: 16478530 DOI: 10.1111/j.1471-4159.2005.03641.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Molecular mechanisms underlying tauopathy remain undetermined. In the current study, single cell gene expression profiling was coupled with custom-designed cDNA array analysis to evaluate tau expression and other cytoskeletal elements within individual neuronal populations in patients with no cognitive impairment (NCI), mild cognitive impairment (MCI), and Alzheimer's disease (AD). Results revealed a shift in the ratio of three-repeat tau (3Rtau) to four-repeat tau (4Rtau) mRNAs within individual human cholinergic basal forebrain (CBF) neurons within nucleus basalis (NB) and CA1 hippocampal neurons during the progression of AD, but not during normal aging. A shift in 3Rtau to 4Rtau may precipitate a cascade of events in the selective vulnerability of neurons, ultimately leading to frank neurofibrillary tangle (NFT) formation in tauopathies including AD.
Collapse
Affiliation(s)
- Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962, USA.
| | | | | | | |
Collapse
|
42
|
Ding ZT, Wang Y, Jiang YP, Yoshida M, Mimuro M, Inagaki T, Iwase T, Hashizume Y. Argyrophilic grain disease: frequency and neuropathology in centenarians. Acta Neuropathol 2006; 111:320-8. [PMID: 16525805 DOI: 10.1007/s00401-006-0043-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2005] [Revised: 12/30/2005] [Accepted: 12/31/2005] [Indexed: 11/26/2022]
Abstract
Argyrophilic grain disease (AGD) is a progressive degenerative disease of the human brain, the prevalence of which increases with advancing age. The features of AGD in autopsied brains from 32 centenarians were studied using phosphorylated tau (AT8) immunostaining combined with Gallyas-Braak staining and 4R tau-specific antibody (RD4) immunostaining. Ten of 32 centenarians were diagnosed as AGD, yielding an overall frequency of 31.3%. In the demented group, nine (39.1%) of 23 cases were found with argyrophilic grains (AGs), while in the non-demented group, AGs were found in only one (11.1%) of nine cases, the difference between them being significant (P<0.05). Among the cases with Alzheimer's disease (AD), five (41.7%) of 12 were found with AGs. One (25%) of four cases with senile dementia with tangles (SDT) also suffered from AGD. Dementia caused by "pure" AGD accounted for 13% (3/23) among demented subjects. Our findings indicated that there is a high frequency of AGD in centenarians. In agreement with previous studies, we favor the view that age may be one of the risk factors for AGD.
Collapse
Affiliation(s)
- Zheng-Tong Ding
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Clinical Features of Argyrophilic Grain Disease: A Retrospective Survey of Cases With Neuropsychiatric Symptoms. Am J Geriatr Psychiatry 2005. [PMID: 16319301 DOI: 10.1097/00019442-200512000-00008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
44
|
Yamazaki M, Hasegawa M, Mori O, Murayama S, Tsuchiya K, Ikeda K, Chen KM, Katayama Y, Oyanagi K. Tau-Positive Fine Granules in the Cerebral White Matter: A Novel Finding Among the Tauopathies Exclusive to Parkinsonism-Dementia Complex of Guam. J Neuropathol Exp Neurol 2005; 64:839-46. [PMID: 16215455 DOI: 10.1097/01.jnen.0000182977.79483.89] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
We examined the autopsied brains of cases of 6 types of tauopathy: parkinsonism-dementia complex of Guam (PDC), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), Pick disease, Alzheimer disease (AD), and myotonic dystrophy together with Guamanian controls. Light microscopy sections of these brains were examined using anti-tau antibodies. Tau-positive fine granules (TFGs) were globe-shaped, and 3 to 6 mum in diameter, were observed predominantly in the frontal white matter in 30 of the 35 patients with PDC. However, no TFGs were found in association with PSP, myotonic dystrophy, Pick disease, AD, or CBD. Western blot analysis of frozen brain tissue taken from the PDC cases revealed that the frontal cortex was hyperphosphorylated and contained 6 tau isoforms (3R+4R tau). However, in the present study, it was revealed that the novel TFGs in the white matter of patients with PDC was composed of 4R tau. Western blot analysis of sarkosyl-insoluble tau from the white matter of the PDC cases showed 2 major bands of 60 and 64 kDa and one minor band of 67 kDa. After dephosphorylation, these bands resolved into one major band of 4-repeat (4R) tau isoform and 3 minor bands of 3-repeat (3R) and 4R tau isoforms. Moreover, the TFGs observed in cases in which the number of neurofibrillary tangles (NFTs) was higher than the threshold level were not correlated with the presence of cortical NFTs. In conclusion, these novel TFGs were found almost exclusively in PDC brains and could therefore be considered as a characteristic neuropathologic marker of this particular tauopathy. The TFGs were hyperphosphorylated tau-positive structures that may be formed by a different mechanism from that used to produce cortical NFTs.
Collapse
Affiliation(s)
- Mineo Yamazaki
- Department of Neuropathology, Tokyo Metropolitan Institute for Neuroscience, Fuchu-shi, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Halverson RA, Lewis J, Frausto S, Hutton M, Muma NA. Tau protein is cross-linked by transglutaminase in P301L tau transgenic mice. J Neurosci 2005; 25:1226-33. [PMID: 15689560 PMCID: PMC6725970 DOI: 10.1523/jneurosci.3263-04.2005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The microtubule-associated protein tau is highly soluble under physiological conditions. However, in tauopathies, tau protein aggregates into insoluble filaments and neurofibrillary tangles (NFTs). The mechanisms underlying the formation of tau filaments and NFTs in tauopathies remain unclear. Several lines of evidence suggest that transglutaminase may cross-link tau into stable, insoluble aggregates, leading to the formation of NFTs in Alzheimer's disease and progressive supranuclear palsy. To further determine the contribution of transglutaminase in the formation of NFTs, we compared the levels of cross-linked tau protein from P301L tau transgenic mice that develop NFTs to four-repeat wild-type (4RWT) tau transgenic and nontransgenic mice that do not develop NFT pathology. Immunoprecipitation and immunoblotting experiments show that transglutaminase cross-links phosphorylated tau in the hindbrain of P301L tau transgenic mice but not in mice overexpressing 4RWT tau and nontransgenic mice. Cross-linked, phosphorylated tau from P301L tau transgenic mice runs as high-molecular mass aggregates on Western blots, similar to cross-linked tau from paired helical filaments of Alzheimer's disease. We also used double-label immunofluorescence to demonstrate colocalization of PHF-1-immunoreactive tau and the transglutaminase-catalyzed cross-link in the hindbrain, spinal cord, and cortex of P301L tau transgenic mice. In the spinal cord, 87% of PHF-1-labeled cells colocalize with the transglutaminase-catalyzed cross-link. Additionally, transglutaminase enzymatic activity is significantly elevated in the spinal cord of P301L tau transgenic mice. These studies further implicate transglutaminase in the formation and/or stabilization of NFT and paired helical filaments and provide a model system to investigate the therapeutic potential of transglutaminase inhibitors in tauopathies.
Collapse
Affiliation(s)
- Robyn A Halverson
- Department of Pharmacology, Loyola University Chicago Medical Center, Maywood, Illinois 60153, USA
| | | | | | | | | |
Collapse
|
46
|
Wenning GK, Jellinger KA. The role of α-synuclein and tau in neurodegenerative movement disorders. Curr Opin Neurol 2005; 18:357-62. [PMID: 16003109 DOI: 10.1097/01.wco.0000168241.53853.32] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Gregor K Wenning
- Department of Neurology, Medical University, Innsbruck, Austria.
| | | |
Collapse
|
47
|
Uchihara T, Tsuchiya K, Nakamura A, Akiyama H. Argyrophilic grains are not always argyrophilic--distinction from neurofibrillary tangles of diffuse neurofibrillary tangles with calcification revealed by comparison between Gallyas and Campbell-Switzer methods. Acta Neuropathol 2005; 110:158-64. [PMID: 15971055 DOI: 10.1007/s00401-005-1031-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 04/13/2005] [Accepted: 04/13/2005] [Indexed: 10/25/2022]
Abstract
Silver staining profiles of argyrophilic grains (AGs) and of neurofibrillary tangles (NFTs) of diffuse neurofibrillary tangles with calcification (DNTC, collectively as DNTC-NFTs) were examined for their relation to tau- and ubiquitin-like immunoreactivity (IR). Pairs of mirror sections were triple-fluorolabeled with an anti-PHF tau (AT8) antibody, an anti-ubiquitin antibody and thiazin red (TR), a fluorochrome that identifies fibrillary structures such as NFTs of Alzheimer's disease (AD). One of the paired sections was subsequently stained with Gallyas method (GAL), and the other with Campbell-Switzer method (CS). Comparison of the same microscopic field on the paired fluorolabeled sections, subsequently silver-stained with either GAL or CS enabled the determination of five different profiles of each structure: AT8-IR, ubiquitin-like-IR, affinity to TR, argyrophilia with GAL or CS staining. AGs, mainly composed of four-repeat (4R) tau, were argyrophilic with GAL but not with CS, and their affinity to TR and ubiquitin-like-IR was not intense. This staining profile of AGs is identical with those of tau-positive structures in the cortex of progressive supranuclear palsy/corticobasal degeneration, both composed of 4R tau. This selective affinity of AGs to GAL is in sharp contrast with Pick bodies, composed of three-repeat (3R) tau, that are positive for CS but not for GAL, as we reported previously. This contrast is explainable if the argyrophilia with CS is related to deposits containing 3R tau, while that with GAL is linked to those containing 4R tau. Indeed, DNTC-NFTs, that contain both 3R and 4R tau, were argyrophilic with CS and GAL, and their affinity to TR and ubiquitin-like-IR were consistent, as we reported previously for NFTs of AD and of Down's syndrome, both similarly composed of 3R and 4R tau. Taken together, differences in molecular composition of tau protein in these deposits are linked to their argyrophilic properties dependent on the staining method in these sporadic tauopathies. Although explanations for these empirical differences are not yet available, awareness of this clear distinction is potentially of diagnostic and pathological significance.
Collapse
Affiliation(s)
- Toshiki Uchihara
- Department of Neuropathology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashi-dai, Fuchu, 183-8526 , Tokyo, Japan.
| | | | | | | |
Collapse
|
48
|
Thal DR, Schultz C, Botez G, Del Tredici K, Mrak RE, Griffin WST, Wiestler OD, Braak H, Ghebremedhin E. The impact of argyrophilic grain disease on the development of dementia and its relationship to concurrent Alzheimer's disease-related pathology. Neuropathol Appl Neurobiol 2005; 31:270-9. [PMID: 15885064 DOI: 10.1111/j.1365-2990.2005.00635.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Argyrophilic grain disease (AGD) constitutes a neurodegenerative disorder that occurs in the brains of the elderly and affects 5% of all patients with dementia. Tau protein-containing lesions known as argyrophilic grains and located predominantly in limbic regions of the brain characterize this disease. Dementia is encountered in only a subset of cases that display the morphological pattern of AGD. The aim of this study is to determine the role of concurrent Alzheimer's disease (AD)-related pathology for the development of dementia in AGD patients. A total of 204 post-mortem brains from 30 demented and 49 nondemented AGD patients, 39 AD patients, and from 86 nondemented controls without AGD were staged for AD-related neurofibrillary tangles (NFTs) as well as amyloid beta-protein (Abeta) deposition. To identify differences in AD-related pathology between demented and nondemented AGD cases, and to differentiate the pattern of AD-related changes in demented and nondemented AGD cases from that seen in AD and nondemented controls, we statistically compared the stages of Abeta and NFT distribution among these groups. Using a logistic regression model, we showed that AGD has a significant effect on the development of dementia beyond that attributable to AD-related pathology (P < 0.005). Demented AGD cases showed lower stages of AD-related pathology than did pure AD cases but higher stages than nondemented AGD patients. AGD associated dementia was seen in the presence of NFT (Braak)-stages II-IV and Abeta-phases 2-3, whereas those stages were not associated with dementia in the absence of AGD. In conclusion, AGD is a clinically relevant neurodegenerative entity that significantly contributes to the development of dementia by lowering the threshold for cognitive deficits in the presence of moderate amounts of AD-related pathology.
Collapse
Affiliation(s)
- D R Thal
- Department of Neuropathology, University of Bonn Medical Center, D-53105 Bonn, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mott RT, Dickson DW, Trojanowski JQ, Zhukareva V, Lee VM, Forman M, Van Deerlin V, Ervin JF, Wang DS, Schmechel DE, Hulette CM. Neuropathologic, biochemical, and molecular characterization of the frontotemporal dementias. J Neuropathol Exp Neurol 2005; 64:420-8. [PMID: 15892300 DOI: 10.1093/jnen/64.5.420] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The frontotemporal dementias (FTDs) are a heterogeneous group of neurodegenerative disorders that are characterized clinically by dementia, personality changes, language impairment, and occasionally extrapyramidal movement disorders. Historically, the diagnosis and classification of FTDs has been fraught with difficulties, especially with regard to establishing a consensus on the neuropathologic diagnosis. Recently, an international group of scientists participated in a consensus conference to develop such neuropathologic criteria. They recommended a diagnostic classification scheme that incorporated a biochemical analysis of the insoluble tau isoform composition, as well as ubiquitin immunohistochemistry. The use and reliability of this classification system has yet to be examined. In this study, we evaluated 21 cases of FTD. Using traditional histochemical stains and tau protein and ubiquitin immunohistochemistry, we separated each case into one of the following categories: classic Pick disease (PiD; n = 7), corticobasal degeneration (CBD; n = 5), dementia lacking distinctive histopathologic features (DLDH; n = 4), progressive supranuclear palsy (PSP; n = 2), frontotemporal lobar degeneration with motor neuron disease or motor neuron disease-type inclusions (FTLD-MND/MNI; n = 2), and neurofibrillary tangle dementia (NFTD; n = 1). Additionally, we independently categorized each case by the insoluble tau isoform pattern, including 3R (n = 5), 4R (n = 7), 3R/4R (n = 3), and no insoluble tau (n = 6). As suggested by the proposed diagnostic scheme, we found that the insoluble tau isoform patterns correlated strongly with the independently derived histopathologic diagnoses (p < 0.001). The data show that cases containing predominantly 3R tau were classic PiD (100%). Cases with predominantly 4R tau were either CBD (71%) or PSP (29%). Cases with both 3R and 4R tau were either a combination of PiD and Alzheimer disease (67%) or NFTD (33%). Finally, cases with no insoluble tau were either DLDH (67%) or FTLD-MND/MNI (33%). To further characterize these cases, we also performed quantitative Western blots for soluble tau, APOE genotyping, and, in selected cases, tau gene sequencing. We show that soluble tau is reduced in DLDH and FTLD-MND/MNI and that APOE4 is overrepresented in PiD and DLDH. We also identified a new family with the R406W mutation and pathology consistent with NFTD. This study validates the recently proposed diagnostic criteria and forms a framework for further refinement of this classification scheme.
Collapse
Affiliation(s)
- Ryan T Mott
- Department of Pathology (Neuropathology), Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fujino Y, Wang DS, Thomas N, Espinoza M, Davies P, Dickson DW. Increased frequency of argyrophilic grain disease in Alzheimer disease with 4R tau-specific immunohistochemistry. J Neuropathol Exp Neurol 2005; 64:209-214. [PMID: 15804052 DOI: 10.1093/jnen/64.3.209] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Argyrophilic grain disease (AGD) is a medial temporal 4R tauopathy with filamentous inclusions in dendrospinal portions of neurons. AGD is often associated with mild Alzheimer-type pathology, but it is difficult to detect AGD in the setting of advanced Alzheimer disease (AD). The frequency of AGD in AD has been difficult to determine because of masking of grains by neurofibrillary lesions. To address this issue, medial temporal lobe sections from AD brains were immunostained with a 4R tau-specific antibody, ET3, which permitted detection of grains even in the setting of advanced neurofibrillary degeneration. AGD was found in 61 of 239 AD cases (26%). The frequency of AGD in AD in this study is higher than in previous studies that relied on less selective staining methods, such as the Gallyas silver stain or immunostaining with phospho-tau antibodies. The frequency of AGD in AD did not correlate with Braak stage or with the density of neurofibrillary tangles and senile plaques in the limbic lobe; however, AD cases with AGD were significantly older than cases without AGD. The MAPT H1 frequency tended to be higher in AD cases with AGD than in those without AGD, but there were no differences in APOE epsilon4 carrier state. These findings suggest advanced age and possibly MAPT H1 are risk factors for AGD, even in the setting of concurrent AD, in which neurofibrillary degeneration is associated with accumulation of both 3R and 4R tau.
Collapse
Affiliation(s)
- Yasuhiro Fujino
- Department of Pathology (Neuropathology), Mayo Clinic College of Medicine, Jacksonville, Florida 32224, USA
| | | | | | | | | | | |
Collapse
|