51
|
Coughlin DG, Litvan I. Progressive supranuclear palsy: Advances in diagnosis and management. Parkinsonism Relat Disord 2020; 73:105-116. [PMID: 32487421 PMCID: PMC7462164 DOI: 10.1016/j.parkreldis.2020.04.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023]
Abstract
Progressive supranuclear palsy (PSP) is a complex clinicopathologic disease with no current cure or disease modulating therapies that can only be definitively confirmed at autopsy. Growing understanding of the phenotypic diversity of PSP has led to expanded clinical criteria and new insights into etiopathogenesis that coupled with improved in vivo biomarkers makes increased access to current clinical trials possible. Current standard-of-care treatment of PSP is multidisciplinary, supportive and symptomatic, and several trials of potentially disease modulating agents have already been completed with disappointing results. Current ongoing clinical trials target the abnormal aggregation of tau through a variety of mechanisms including immunotherapy and gene therapy offer a more direct method of treatment. Here we review PSP clinicopathologic correlations, in vivo biomarkers including MRI, PET, and CSF biomarkers. We additionally review current pharmacologic and non-pharmacologic methods of treatment, prior and ongoing clinical trials in PSP. Newly expanded clinical criteria and improved specific biomarkers will aid in identifying patients with PSP earlier and more accurately and expand access to these potentially beneficial clinical trials.
Collapse
Affiliation(s)
- David G Coughlin
- Department of Neurosciences, University of California San Diego, San Diego, CA, 92093, USA
| | - Irene Litvan
- Department of Neurosciences, University of California San Diego, San Diego, CA, 92093, USA.
| |
Collapse
|
52
|
Davidowitz EJ, Krishnamurthy PK, Lopez P, Jimenez H, Adrien L, Davies P, Moe JG. In Vivo Validation of a Small Molecule Inhibitor of Tau Self-Association in htau Mice. J Alzheimers Dis 2020; 73:147-161. [PMID: 31771053 PMCID: PMC6957711 DOI: 10.3233/jad-190465] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 01/27/2023]
Abstract
Tau oligomers have been shown to transmit tau pathology from diseased neurons to healthy neurons through seeding, tau misfolding, and aggregation that is thought to play an influential role in the progression of Alzheimer's disease (AD) and related tauopathies. To develop a small molecule therapeutic for AD and related tauopathies, we have developed in vitro and cellular assays to select molecules inhibiting the first step in tau aggregation, the self-association of tau into oligomers. In vivo validation studies of an optimized lead compound were independently performed in the htau mouse model of tauopathy that expresses the human isoforms of tau without inherited tauopathy mutations that are irrelevant to AD. Treated mice did not show any adverse events related to the compound. The lead compound significantly reduced the level of self-associated tau and total and phosphorylated insoluble tau aggregates. The dose response was linear with respect to levels of compound in the brain. A confirmatory study was performed with male htau mice that gave consistent results. The results validated our screening approach by showing that targeting tau self-association can inhibit the entire tau aggregation pathway by using the selected and optimized lead compound whose activity translated from in vitro and cellular assays to an in vivo model of tau aggregation.
Collapse
Affiliation(s)
| | | | | | - Heidy Jimenez
- The Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Leslie Adrien
- The Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Peter Davies
- The Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | | |
Collapse
|
53
|
Reelin reverts biochemical, physiological and cognitive alterations in mouse models of Tauopathy. Prog Neurobiol 2019; 186:101743. [PMID: 31870804 DOI: 10.1016/j.pneurobio.2019.101743] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 10/24/2019] [Accepted: 12/18/2019] [Indexed: 01/13/2023]
Abstract
Reelin is an extracellular protein crucial for adult brain plasticity. Moreover, Reelin is protective against amyloid-β (Aβ) pathology in Alzheimer's Disease (AD), reducing plaque deposition, synaptic loss and cognitive decline. Given that Tau protein plays a key role in AD pathogenesis, and that the Reelin pathway modulates Tau phosphorylation, here we explored the involvement of Reelin in AD-related Tau pathology. We found that Reelin overexpression modulates the levels of Tau phosphorylation in AD-related epitopes in VLW mice expressing human mutant Tau. in vitro, Reelin reduced the Aβ-induced missorting of axonal Tau and neurofilament proteins to dendrites. Reelin also reverted in vivo the toxic somatodendritic localization of phosphorylated Tau. Finally, overexpression of Reelin in VLW mice improved long-term potentiation and long-term memory cognitive performance thus masking the cognitive and physiological deficits in VLW mice. These data suggest that the Reelin pathway, which is also protective against Aβ pathology, modulates fundamental traits of Tau pathology, strengthening the potential of Reelin as a therapeutic target in AD.
Collapse
|
54
|
Liu Y, Wei W, Baazaoui N, Liu F, Iqbal K. Inhibition of AMD-Like Pathology With a Neurotrophic Compound in Aged Rats and 3xTg-AD Mice. Front Aging Neurosci 2019; 11:309. [PMID: 31803044 PMCID: PMC6877482 DOI: 10.3389/fnagi.2019.00309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 10/28/2019] [Indexed: 11/17/2022] Open
Abstract
Age-associated macular degeneration (AMD), which leads to loss of vision at its end stage, is one of the most common neurodegenerative diseases among the elderly. However, to date, no effective drug therapy is available for the prevention of AMD. Here, we report the occurrence of AMD pathology and its prevention by chronic treatment with the neurotrophic peptidergic compound P021, in aged rats and 3xTg-AD mice. We found photoreceptor degeneration, lipofuscin granules, vacuoles, and atrophy in retinal pigment epithelium (RPE) as well as Bruch’s membrane (BM) thickening; in aged rats, we even found rosette-like structure formation. Microgliosis and astrogliosis were observed in different retinal layers. In addition, we also found that total tau, phosphorylated tau, Aβ/APP, and VEGF were widely distributed in the sub-retina of aged rats and 3xTg mice. Importantly, chronic treatment with P021 for 3 months in rats and for 18 months in 3xTg mice ameliorated the pathological changes above. These findings indicate the therapeutic potential of P021 for prevention and treatment of AMD and retinal changes associated with aging and Alzheimer’s disease.
Collapse
Affiliation(s)
- Yinghua Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States.,Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Clinical Pharmacology, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wei Wei
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States.,Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, China
| | - Narjes Baazaoui
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| |
Collapse
|
55
|
Li D, Cho YK. High specificity of widely used phospho-tau antibodies validated using a quantitative whole-cell based assay. J Neurochem 2019; 152:122-135. [PMID: 31325178 DOI: 10.1111/jnc.14830] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/25/2019] [Accepted: 07/11/2019] [Indexed: 02/04/2023]
Abstract
Antibodies raised against defined phosphorylation sites of the microtubule-associated protein tau are widely used in scientific research and being applied in clinical assays. However, recent studies have revealed an alarming degree of non-specific binding found in these antibodies. In order to quantify and compare the specificity phospho-tau antibodies and other post-translational modification site-specific antibodies in general, a measure of specificity is urgently needed. Here, we report a robust flow cytometry assay using human embryonic kidney cells that enables the determination of a specificity parameter termed Φ, which measures the fraction of non-specific signal in antibody binding. We validate our assay using anti-tau antibodies with known specificity profiles, and apply it to measure the specificity of seven widely used phospho-tau antibodies (AT270, AT8, AT100, AT180, PHF-6, TG-3, and PHF-1) among others. We successfully determined the Φ values for all antibodies except AT100, which did not show detectable binding in our assay. Our results show that antibodies AT8, AT180, PHF-6, TG-3, and PHF-1 have Φ values near 1, which indicates no detectable non-specific binding. AT270 showed Φ value around 0.8, meaning that approximately 20% of the binding signal originates from non-specific binding. Further analyses using immunocytochemistry and western blotting confirmed the presence of non-specific binding of AT270 to non-tau proteins found in human embryonic kidney cells and the mouse hippocampus. We anticipate that the quantitative approach and parameter introduced here will be widely adopted as a standard for reporting the specificity for phospho-tau antibodies, and potentially for post-translational modification targeting antibodies in general. Cover Image for this issue: doi: 10.1111/jnc.14727.
Collapse
Affiliation(s)
- Dan Li
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Yong Ku Cho
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut, USA.,Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, USA.,Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
56
|
Vandermeeren M, Borgers M, Van Kolen K, Theunis C, Vasconcelos B, Bottelbergs A, Wintmolders C, Daneels G, Willems R, Dockx K, Delbroek L, Marreiro A, Ver Donck L, Sousa C, Nanjunda R, Lacy E, Van De Casteele T, Van Dam D, De Deyn PP, Kemp JA, Malia TJ, Mercken MH. Anti-Tau Monoclonal Antibodies Derived from Soluble and Filamentous Tau Show Diverse Functional Properties in vitro and in vivo. J Alzheimers Dis 2019; 65:265-281. [PMID: 30040731 DOI: 10.3233/jad-180404] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The tau spreading hypothesis provides rationale for passive immunization with an anti-tau monoclonal antibody to block seeding by extracellular tau aggregates as a disease-modifying strategy for the treatment of Alzheimer's disease (AD) and potentially other tauopathies. As the biochemical and biophysical properties of the tau species responsible for the spatio-temporal sequences of seeding events are poorly defined, it is not yet clear which epitope is preferred for obtaining optimal therapeutic efficacy. Our internal tau antibody collection has been generated by immunizations with different tau species: aggregated- and non-aggregated tau and human postmortem AD brain-derived tau fibrils. In this communication, we describe and characterize a set of these anti-tau antibodies for their biochemical and biophysical properties, including binding, tissue staining by immunohistochemistry, and epitope. The antibodies bound to different domains of the tau protein and some were demonstrated to be isoform-selective (PT18 and hTau56) or phospho-selective (PT84). Evaluation of the antibodies in cellular- and in vivo seeding assays revealed clear differences in maximal efficacy. Limited proteolysis experiments support the hypothesis that some epitopes are more exposed than others in the tau seeds. Moreover, antibody efficacy seems to depend on the structural properties of fibrils purified from tau Tg mice- and postmortem human AD brain.
Collapse
Affiliation(s)
- Marc Vandermeeren
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Marianne Borgers
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Kristof Van Kolen
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Clara Theunis
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Bruno Vasconcelos
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Astrid Bottelbergs
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Cindy Wintmolders
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Guy Daneels
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Roland Willems
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Koen Dockx
- Discovery Sciences, Janssen Research and Development, Beerse, Belgium
| | - Lore Delbroek
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - André Marreiro
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Luc Ver Donck
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Cristiano Sousa
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Rupesh Nanjunda
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Eilyn Lacy
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | | | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Alzheimer Research Center, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Peter Paul De Deyn
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Alzheimer Research Center, University Medical Center Groningen (UMCG), Groningen, The Netherlands.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.,Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - John A Kemp
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium.,Syndesi therapeutics, Belgium
| | - Thomas J Malia
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Marc H Mercken
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| |
Collapse
|
57
|
Abner EL, Neltner JH, Jicha GA, Patel E, Anderson SL, Wilcock DM, Van Eldik LJ, Nelson PT. Diffuse Amyloid-β Plaques, Neurofibrillary Tangles, and the Impact of APOE in Elderly Persons' Brains Lacking Neuritic Amyloid Plaques. J Alzheimers Dis 2019; 64:1307-1324. [PMID: 30040735 DOI: 10.3233/jad-180514] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Data from a large autopsy series were analyzed to address questions pertinent to primary age-related tauopathy (PART) and Alzheimer's disease (AD): what factors are associated with increased severity of neurofibrillary degeneration in brains that lack neuritic amyloid plaques?; is there an association between Apolipoprotein E (APOE) alleles and PART pathologic severity independent of amyloid-β (Aβ) deposits?; and, how do the stains used to detect plaques and tangles impact the experimental results? Neuropathologic data were evaluated from elderly research volunteers whose brain autopsies were performed at University of Kentucky Alzheimer's Disease Center (UK-ADC; N = 145 subjects). All of the included subjects' brains lacked neuritic amyloid plaques according to the CERAD diagnostic criteria and the average final MMSE score before death was 26.8±4.6 stdev. The study incorporated evaluation of tissue with both silver histochemical stains and immunohistochemical stains to compare results; the immunohistochemical stains (Aβ and phospho-tau) were scanned and quantified using digital pathologic methods. Immunohistochemical stains provided important advantages over histochemical stains due to sensitivity and detectability via digital methods. When AD-type pathology was in its presumed earliest phases, neocortical parenchymal Aβ deposits were associated with increased medial temporal lobe neurofibrillary tangles. The observation supports the NIA-AA consensus recommendation for neuropathologic diagnoses, because even these "diffuse" Aβ deposits signal that AD pathobiologic mechanisms are occurring. Further, the data were most compatible with the hypothesis that the APOEɛ4 allele exerts its effect(s) via driving Aβ deposition, i.e., an "upstream" influence, rather than being associated directly with Aβ- independent PART pathology.
Collapse
Affiliation(s)
- Erin L Abner
- Department of Epidemiology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Janna H Neltner
- Department of Pathology, Division of Neuropathology, University of Kentucky, Lexington, KY, USA
| | - Gregory A Jicha
- Department of Neurology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Ela Patel
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Sonya L Anderson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Donna M Wilcock
- Department of Physiology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Linda J Van Eldik
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Department of Pathology, Division of Neuropathology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
58
|
Naseri NN, Wang H, Guo J, Sharma M, Luo W. The complexity of tau in Alzheimer's disease. Neurosci Lett 2019; 705:183-194. [PMID: 31028844 PMCID: PMC7060758 DOI: 10.1016/j.neulet.2019.04.022] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/14/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is characterized by two major pathological lesions in the brain, amyloid plaques and neurofibrillary tangles (NFTs) composed mainly of amyloid-β (Aβ) peptides and hyperphosphorylated tau, respectively. Although accumulation of toxic Aβ species in the brain has been proposed as one of the important early events in AD, continued lack of success of clinical trials based on Aβ-targeting drugs has triggered the field to seek out alternative disease mechanisms and related therapeutic strategies. One of the new approaches is to uncover novel roles of pathological tau during disease progression. This review will primarily focus on recent advances in understanding the contributions of tau to AD.
Collapse
Affiliation(s)
- Nima N Naseri
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| | - Hong Wang
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, USA
| | - Jennifer Guo
- The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
59
|
Gibbons GS, Kim SJ, Robinson JL, Changolkar L, Irwin DJ, Shaw LM, Lee VMY, Trojanowski JQ. Detection of Alzheimer's disease (AD) specific tau pathology with conformation-selective anti-tau monoclonal antibody in co-morbid frontotemporal lobar degeneration-tau (FTLD-tau). Acta Neuropathol Commun 2019; 7:34. [PMID: 30832741 PMCID: PMC6399892 DOI: 10.1186/s40478-019-0687-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/23/2019] [Indexed: 12/12/2022] Open
Abstract
Pathological tau aggregates in Alzheimer’s disease (AD) and frontotemporal lobar degeneration-tau (FTLD-tau) adopt distinct conformations differentiated by the AD-tau specific monoclonal antibody (mAb) GT-38 that are not readily visualized using phosphorylation-specific anti-tau mAbs. To determine the extent of co-morbid AD-tau pathology in FTLD-tau, we performed immunohistochemical (IHC) staining with GT-38 and assigned Braak stages of AD-tau in a cohort 180 FTLD-tau cases consisting of corticobasal degeneration (CBD; n = 49), progressive supranuclear palsy (PSP; n = 109), and Pick’s disease (PiD; n = 22). Nearly two-thirds of patients (n = 115 of 180, 63.8%) with FTLD-tau had some degree of comorbid AD-tau pathology and 20.5% of the FTLD-tau cohort had Braak stage ≥B2, consistent with medium-to-high-level AD neuropathological change (ADNPC). The PSP group had the highest frequency of medium-high AD-tau pathology compared to other tauopathies (PSP = 31/109, 28.4%; Picks = 2/22, 9.1%, CBD = 4/49, 8.2%) but neuropathological diagnosis was not found to be a significant independent predictor of medium-high AD Braak stage in a multivariate model after accounting for age at death (OR = 1.09; 95% CI = 1.03–1.15; p = 0.002) and CERAD plaque scores (OR = 3.75, 95% CI = 1.58–8.89; p = 0.003), suggesting there is no predilection for a specific FTLD tauopathy to develop AD-tau co-pathology after accounting for age. Patients with FTLD-tau who had, clinically significant, medium-high AD-tau pathology had significantly higher antemortem CSF levels of both total-tau (t-tau; mean = 89.98 pg/ml, SD = 36.70 pg/ml) and phosphorylated-tau (p-tau; mean = 20.45 pg/ml, SD = 9.31 pg/ml) compared to patients with negligible-low AD-tau, t-tau (mean = 43.04 pg/ml, SD = 25.40 pg/ml) and p-tau (mean = 11.90 pg/ml, SD = 4.48 pg/ml) (p ≤ 0.001 both). Finally, in an exploratory analysis in our largest pathology group (PSP) we find an association of GT-38 AD-tau Braak stage with lower baseline MMSE (p = 0.03). Together, these finding validate the use of GT-38 to selectively detect AD-tau pathology in the context of FTLD-tau and provides a novel tool to investigate associations of clinical phenotypes amongst co-morbid tauopathies.
Collapse
|
60
|
Bignante EA, Ponce NE, Heredia F, Musso J, Krawczyk MC, Millán J, Pigino GF, Inestrosa NC, Boccia MM, Lorenzo A. APP/Go protein Gβγ-complex signaling mediates Aβ degeneration and cognitive impairment in Alzheimer's disease models. Neurobiol Aging 2017; 64:44-57. [PMID: 29331876 DOI: 10.1016/j.neurobiolaging.2017.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/05/2017] [Accepted: 12/10/2017] [Indexed: 11/15/2022]
Abstract
Deposition of amyloid-β (Aβ), the proteolytic product of the amyloid precursor protein (APP), might cause neurodegeneration and cognitive decline in Alzheimer's disease (AD). However, the direct involvement of APP in the mechanism of Aβ-induced degeneration in AD remains on debate. Here, we analyzed the interaction of APP with heterotrimeric Go protein in primary hippocampal cultures and found that Aβ deposition dramatically enhanced APP-Go protein interaction in dystrophic neurites. APP overexpression rendered neurons vulnerable to Aβ toxicity by a mechanism that required Go-Gβγ complex signaling and p38-mitogen-activated protein kinase activation. Gallein, a selective pharmacological inhibitor of Gβγ complex, inhibited Aβ-induced dendritic and axonal dystrophy, abnormal tau phosphorylation, synaptic loss, and neuronal cell death in hippocampal neurons expressing endogenous protein levels. In the 3xTg-AD mice, intrahippocampal application of gallein reversed memory impairment associated with early Aβ pathology. Our data provide further evidence for the involvement of APP/Go protein in Aβ-induced degeneration and reveal that Gβγ complex is a signaling target potentially relevant for developing therapies for halting Aβ degeneration in AD.
Collapse
Affiliation(s)
- Elena Anahi Bignante
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina; Instituto Universitario de Ciencias Biomédicas de Córdoda (IUCBC), Argentina
| | - Nicolás Eric Ponce
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florencia Heredia
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Juliana Musso
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María C Krawczyk
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julieta Millán
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gustavo F Pigino
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Punta Arenas, Chile
| | - Mariano M Boccia
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alfredo Lorenzo
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
61
|
Chiasseu M, Alarcon-Martinez L, Belforte N, Quintero H, Dotigny F, Destroismaisons L, Vande Velde C, Panayi F, Louis C, Di Polo A. Tau accumulation in the retina promotes early neuronal dysfunction and precedes brain pathology in a mouse model of Alzheimer's disease. Mol Neurodegener 2017; 12:58. [PMID: 28774322 PMCID: PMC5543446 DOI: 10.1186/s13024-017-0199-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/31/2017] [Indexed: 12/14/2022] Open
Abstract
Background Tau is an axon-enriched protein that binds to and stabilizes microtubules, and hence plays a crucial role in neuronal function. In Alzheimer’s disease (AD), pathological tau accumulation correlates with cognitive decline. Substantial visual deficits are found in individuals affected by AD including a preferential loss of retinal ganglion cells (RGCs), the neurons that convey visual information from the retina to the brain. At present, however, the mechanisms that underlie vision changes in these patients are poorly understood. Here, we asked whether tau plays a role in early retinal pathology and neuronal dysfunction in AD. Methods Alterations in tau protein and gene expression, phosphorylation, and localization were investigated by western blots, qPCR, and immunohistochemistry in the retina and visual pathways of triple transgenic mice (3xTg) harboring mutations in the genes encoding presenilin 1 (PS1M146 V), amyloid precursor protein (APPSwe), and tau (MAPTP301L). Anterograde axonal transport was assessed by intraocular injection of the cholera toxin beta subunit followed by quantification of tracer accumulation in the contralateral superior colliculus. RGC survival was analyzed on whole-mounted retinas using cell-specific markers. Reduction of tau expression was achieved following intravitreal injection of targeted siRNA. Results Our data demonstrate an age-related increase in endogenous retinal tau characterized by epitope-specific hypo- and hyper-phosphorylation in 3xTg mice. Retinal tau accumulation was observed as early as three months of age, prior to the reported onset of behavioral deficits, and preceded tau aggregation in the brain. Intriguingly, tau build up occurred in RGC soma and dendrites, while tau in RGC axons in the optic nerve was depleted. Tau phosphorylation changes and missorting correlated with substantial defects in anterograde axonal transport that preceded RGC death. Importantly, targeted siRNA-mediated knockdown of endogenous tau improved anterograde transport along RGC axons. Conclusions Our study reveals profound tau pathology in the visual system leading to early retinal neuron damage in a mouse model of AD. Importantly, we show that tau accumulation promotes anterograde axonal transport impairment in vivo, and identify this response as an early feature of neuronal dysfunction that precedes cell death in the AD retina. These findings provide the first proof-of-concept that a global strategy to reduce tau accumulation is beneficial to improve axonal transport and mitigate functional deficits in AD and tauopathies.
Collapse
Affiliation(s)
- Marius Chiasseu
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Nicolas Belforte
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Heberto Quintero
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Florence Dotigny
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Laurie Destroismaisons
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Christine Vande Velde
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Fany Panayi
- Institut de Recherches Servier, 78290, Croissy-sur-Seine, France
| | - Caroline Louis
- Institut de Recherches Servier, 78290, Croissy-sur-Seine, France
| | - Adriana Di Polo
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada.
| |
Collapse
|
62
|
KrishnaKumar VG, Gupta S. Simplified method to obtain enhanced expression of tau protein from E. coli and one-step purification by direct boiling. Prep Biochem Biotechnol 2017; 47:530-538. [DOI: 10.1080/10826068.2016.1275012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- V. Guru KrishnaKumar
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, India
| | - Sharad Gupta
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, India
| |
Collapse
|
63
|
Swanson E, Breckenridge L, McMahon L, Som S, McConnell I, Bloom GS. Extracellular Tau Oligomers Induce Invasion of Endogenous Tau into the Somatodendritic Compartment and Axonal Transport Dysfunction. J Alzheimers Dis 2017; 58:803-820. [PMID: 28482642 PMCID: PMC5581403 DOI: 10.3233/jad-170168] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aggregates composed of the microtubule associated protein, tau, are a hallmark of Alzheimer's disease and non-Alzheimer's tauopathies. Extracellular tau can induce the accumulation and aggregation of intracellular tau, and tau pathology can be transmitted along neural networks over time. There are six splice variants of central nervous system tau, and various oligomeric and fibrillar forms are associated with neurodegeneration in vivo. The particular extracellular forms of tau capable of transferring tau pathology from neuron to neuron remain ill defined, however, as do the consequences of intracellular tau aggregation on neuronal physiology. The present study was undertaken to compare the effects of extracellular tau monomers, oligomers, and filaments comprising various tau isoforms on the behavior of cultured neurons. We found that 2N4R or 2N3R tau oligomers provoked aggregation of endogenous intracellular tau much more effectively than monomers or fibrils, or of oligomers made from other tau isoforms, and that a mixture of all six isoforms most potently provoked intracellular tau accumulation. These effects were associated with invasion of tau into the somatodendritic compartment. Finally, we observed that 2N4R oligomers perturbed fast axonal transport of membranous organelles along microtubules. Intracellular tau accumulation was often accompanied by increases in the run length, run time and instantaneous velocity of membranous cargo. This work indicates that extracellular tau oligomers can disrupt normal neuronal homeostasis by triggering axonal tau accumulation and loss of the polarized distribution of tau, and by impairing fast axonal transport.
Collapse
Affiliation(s)
- Eric Swanson
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | | | - Lloyd McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Sreemoyee Som
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Ian McConnell
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - George S. Bloom
- Department of Biology, University of Virginia, Charlottesville, VA, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
64
|
Arendt T, Stieler JT, Holzer M. Tau and tauopathies. Brain Res Bull 2016; 126:238-292. [DOI: 10.1016/j.brainresbull.2016.08.018] [Citation(s) in RCA: 333] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022]
|
65
|
Bolton Hall AN, Joseph B, Brelsfoard JM, Saatman KE. Repeated Closed Head Injury in Mice Results in Sustained Motor and Memory Deficits and Chronic Cellular Changes. PLoS One 2016; 11:e0159442. [PMID: 27427961 PMCID: PMC4948770 DOI: 10.1371/journal.pone.0159442] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/17/2016] [Indexed: 02/07/2023] Open
Abstract
Millions of mild traumatic brain injuries (TBIs) occur every year in the United States, with many people subject to multiple head injuries that can lead to chronic behavioral dysfunction. We previously reported that mild TBI induced using closed head injuries (CHI) repeated at 24h intervals produced more acute neuron death and glial reactivity than a single CHI, and increasing the length of time between injuries to 48h reduced the cumulative acute effects of repeated CHI. To determine whether repeated CHI is associated with behavioral dysfunction or persistent cellular damage, mice receiving either five CHI at 24h intervals, five CHI at 48h intervals, or five sham injuries at 24h intervals were evaluated across a 10 week period after injury. Animals with repeated CHI exhibited motor coordination and memory deficits, but not gait abnormalities when compared to sham animals. At 10wks post-injury, no notable neuron loss or glial reactivity was observed in the cortex, hippocampus, or corpus callosum. Argyrophilic axons were found in the pyramidal tract of some injured animals, but neither silver stain accumulation nor inflammatory responses in the injury groups were statistically different from the sham group in this region. However, argyrophilic axons, microgliosis and astrogliosis were significantly increased within the optic tract of injured animals. Repeated mild CHI also resulted in microgliosis and a loss of neurofilament protein 200 in the optic nerve. Lengthening the inter-injury interval from 24h to 48h did not effectively reduce these behavioral or cellular responses. These results suggest that repeated mild CHI results in persistent behavioral dysfunction and chronic pathological changes within the visual system, neither of which was significantly attenuated by lengthening the inter-injury interval from 24h to 48h.
Collapse
Affiliation(s)
- Amanda N. Bolton Hall
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Binoy Joseph
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jennifer M. Brelsfoard
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| |
Collapse
|
66
|
Abstract
Abundant neurofibrillary lesions in certain brain regions constitute one of the defining neuropathological characteristics of Alzheimer's disease, where their presence correlates with the degree of dementia. An understanding of the mechanisms that lead to the neurofibrillary pathology is critical for elucidating the pathogenesis of Alzheimer's disease and for developing effective therapeutic strategies. Neurofibrillary lesions consist of neurofibrillary tangles, neuropil threads, and abnormal neurites. Ultrastructurally, each of these lesions consists of abnormal paired helical and straight filaments. These filaments are made of the six brain isoforms of microtubule-associated protein tau in a hyperphosphorylated and an abnormally phosphorylated state. Several candidate protein kinases and protein phosphatases for the hyperphos phorylation of tau have been identified. Moreover, recent results suggest that an interaction between tau protein and sulfated glycosaminoglycans may play an important role in inducing both the hyperphosphor ylation of tau and the formation of paired helical and straight filaments. NEUROSCIENTIST 3:131-141, 1997
Collapse
Affiliation(s)
- Michel Goedert
- Medical Research Council Laboratory of Molecular Biology Cambridge
| |
Collapse
|
67
|
Combs B, Hamel C, Kanaan NM. Pathological conformations involving the amino terminus of tau occur early in Alzheimer's disease and are differentially detected by monoclonal antibodies. Neurobiol Dis 2016; 94:18-31. [PMID: 27260838 PMCID: PMC4983528 DOI: 10.1016/j.nbd.2016.05.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/10/2016] [Accepted: 05/30/2016] [Indexed: 01/31/2023] Open
Abstract
Conformational changes involving the amino terminus of the tau protein are among the earliest alterations associated with tau pathology in Alzheimer’s disease and other tauopathies. This region of tau contains a phosphatase-activating domain (PAD) that is aberrantly exposed in pathological forms of the protein, an event that is associated with disruptions in anterograde fast axonal transport. We utilized four antibodies that recognize the amino terminus of tau, TNT1, TNT2 (a novel antibody), Tau12, and Tau13, to further study this important region. Using scanning alanine mutations in recombinant tau proteins, we refined the epitopes of each antibody. We examined the antibodies’ relative abilities to specifically label pathological tau in non-denaturing and denaturing assays to gain insight into some of the mechanistic details of PAD exposure. We then determined the pattern of tau pathology labeled by each antibody in human hippocampal sections at various disease stages in order to characterize PAD exposure in the context of disease progression. The characteristics of reactivity for the antibodies fell into two groups. TNT1 and TNT2 recognized epitopes within amino acids 7–12 and specifically identified recombinant tau aggregates and pathological tau from Alzheimer’s disease brains in a conformation-dependent manner. These antibodies labeled early pre-tangle pathology from neurons in early Braak stages and colocalized with thiazine red, a marker of fibrillar pathology, in classic neurofibrillary tangles. However, late tangles were negative for TNT1 and TNT2 indicating a loss of the epitope in later stages of tangle evolution. In contrast, Tau12 and Tau13 both identified discontinuous epitopes in the amino terminus and were unable to differentiate between normal and pathological tau in biochemical and tissue immunohistological assays. Despite the close proximity of these epitopes, the antibodies demonstrated remarkably different abilities to identify pathological changes in tau indicating that detection of conformational alterations involving PAD exposure is not achieved by all N-terminal tau antibodies and that a relatively discrete region of the N-terminus (i.e., amino acids 7–12, the TNT1 and TNT2 epitope) is central to the differences between normal and pathological tau. The appearance of PAD in early tau pathology and its disappearance in late-stage tangles suggest that toxic forms of tau are associated with the earliest forms of tau deposits. Collectively, these findings demonstrate that the TNT antibodies are useful markers for early conformational display of PAD and provide information regarding conformational changes that have potential implications in the toxic mechanisms of tau pathology.
Collapse
Affiliation(s)
- Benjamin Combs
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA.
| | - Chelsey Hamel
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA.
| | - Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA; Neuroscience Program, Michigan State University, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary's, 220 Cherry St SE, Grand Rapids, MI 49503, USA.
| |
Collapse
|
68
|
Day RJ, McCarty KL, Ockerse KE, Head E, Rohn TT. Proteolytic Cleavage of Apolipoprotein E in the Down Syndrome Brain. Aging Dis 2016; 7:267-77. [PMID: 27330841 PMCID: PMC4898923 DOI: 10.14336/ad.2015.1020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/20/2015] [Indexed: 11/17/2022] Open
Abstract
Down syndrome (DS) is one of the most common genetic causes of intellectual disability and is characterized by a number of behavioral as well as cognitive symptoms. Many of the neuropathological features of early-onset Alzheimer’s disease (AD) including senile plaques and neurofibrillary tangles (NFTs) are also present in people with DS as a result of triplication of the amyloid precursor gene on chromosome 21. Evidence suggests that harboring one or both apolipoprotein E4 (APOE4) alleles may increase the risk for AD due to the proteolytic cleavage of apoE4 and a subsequent loss of function. To investigate a role for the apoE proteolysis in vivo, we compared three autopsy groups; 7 DS with AD neuropathology cases over 40 years, 5 young DS cases without AD pathology under 40 years (YDS) and 5 age-matched control cases over 40 years by immunohistochemistry utilizing an antibody that detects the amino-terminal fragment of apoE. Application of this antibody, termed the amino-terminal apoE fragment antibody (nApoECF) revealed labeling of pyramidal neurons in the frontal cortex of YDS cases, whereas in the DS-AD group, labeling with nApoECF was prominent within NFTs. NFT labeling with nApoECF was significantly greater in the hippocampus versus the frontal cortex in the same DS-AD cases, suggesting a regional distribution of truncated apoE. Colocalization immunofluorescence experiments indicated that 52.5% and 53.2% of AT8- and PHF-1-positive NFTs, respectively, also contained nApoECF. Collectively, these data support a role for the proteolytic cleavage of apoE in DS and suggest that apoE fragmentation is closely associated with NFTs.
Collapse
Affiliation(s)
- Ryan J Day
- 1Department of Biological Sciences, Boise State University, Boise, Idaho, 83725, USA
| | - Katie L McCarty
- 2Department of Pharmacology& Nutritional Sciences, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Kayla E Ockerse
- 1Department of Biological Sciences, Boise State University, Boise, Idaho, 83725, USA
| | - Elizabeth Head
- 2Department of Pharmacology& Nutritional Sciences, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Troy T Rohn
- 1Department of Biological Sciences, Boise State University, Boise, Idaho, 83725, USA
| |
Collapse
|
69
|
Domise M, Didier S, Marinangeli C, Zhao H, Chandakkar P, Buée L, Viollet B, Davies P, Marambaud P, Vingtdeux V. AMP-activated protein kinase modulates tau phosphorylation and tau pathology in vivo. Sci Rep 2016; 6:26758. [PMID: 27230293 PMCID: PMC4882625 DOI: 10.1038/srep26758] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/04/2016] [Indexed: 12/12/2022] Open
Abstract
Neurofibrillary tangles (NFTs) are the pathological hallmark of neurodegenerative diseases commonly known as tauopathies. NFTs result from the intracellular aggregation of abnormally and hyperphosphorylated tau proteins. Tau functions, which include the regulation of microtubules dynamics, are dependent on its phosphorylation status. As a consequence, any changes in tau phosphorylation can have major impacts on synaptic plasticity and memory. Recently, it has been demonstrated that AMP-activated protein kinase (AMPK) was deregulated in the brain of Alzheimer's disease (AD) patients where it co-localized with phosphorylated tau in pre-tangle and tangle-bearing neurons. Besides, it was found that AMPK was a tau kinase in vitro. Here, we find that endogenous AMPK activation in mouse primary neurons induced an increase of tau phosphorylation at multiple sites, whereas AMPK inhibition led to a rapid decrease of tau phosphorylation. We further show that AMPK mice deficient for one of the catalytic alpha subunits displayed reduced endogenous tau phosphorylation. Finally, we found that AMPK deficiency reduced tau pathology in the PS19 mouse model of tauopathy. These results show that AMPK regulates tau phosphorylation in mouse primary neurons as well as in vivo, and thus suggest that AMPK could be a key player in the development of AD pathology.
Collapse
Affiliation(s)
- Manon Domise
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT, F-59000 Lille, France
| | - Sébastien Didier
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT, F-59000 Lille, France
| | - Claudia Marinangeli
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT, F-59000 Lille, France
| | - Haitian Zhao
- Litwin-Zucker Research Center for the Study of Alzheimer's disease, The Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | - Pallavi Chandakkar
- Litwin-Zucker Research Center for the Study of Alzheimer's disease, The Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT, F-59000 Lille, France
| | - Benoit Viollet
- Institut Cochin, Inserm U1016, Paris 75014, France.,CNRS, UMR 8104, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Peter Davies
- Litwin-Zucker Research Center for the Study of Alzheimer's disease, The Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | - Philippe Marambaud
- Litwin-Zucker Research Center for the Study of Alzheimer's disease, The Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | - Valérie Vingtdeux
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT, F-59000 Lille, France
| |
Collapse
|
70
|
Critical residues involved in tau binding to fyn: implications for tau phosphorylation in Alzheimer's disease. Acta Neuropathol Commun 2016; 4:49. [PMID: 27193083 PMCID: PMC4870772 DOI: 10.1186/s40478-016-0317-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/26/2016] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterised by neuropathological deposits of amyloid plaques and neurofibrillary tangles comprised of β-amyloid and tau protein, respectively. In AD, tau becomes abnormally phosphorylated and aggregates to form intracellular deposits. However, the mechanisms by which tau exerts neurotoxicity in disease remain unclear. Recent studies have suggested that the presence of tau at synapses may indicate a role in neuronal signalling, which could be disrupted in pathological conditions. The non-receptor-associated tyrosine kinase fyn is located at the dendrite in neurons, where it was recently shown to interact with tau to stabilise receptor complexes at the post-synaptic density. Fyn also co-localises with tau in a proportion of neurons containing tau tangles in AD and fyn is also a tau kinase. Hence, tau-fyn interactions could play a pathogenic role in AD. Here we report the identification of critical proline residues, Pro213, Pro216, and Pro219, located within the fifth and sixth Pro-X-X-Pro motifs in the proline-rich region of tau, that are important for its binding to fyn. These residues in tau are flanked by numerous phosphorylation sites and therefore we investigated the relationship between fyn and the degree of tau phosphorylation in human post-mortem brain tissue. We found no difference in the amount of fyn present in control and AD brain. Notably, however, there was a significant correlation between fyn and phosphorylated tau at specific phospho-epitopes in control, but not in AD brain. Our results suggest that the pathological mechanisms underlying AD, that result in increased tau phosphorylation, may disrupt the physiological relationship between tau phosphorylation and fyn.
Collapse
|
71
|
Intranasal Insulin Prevents Anesthesia-Induced Spatial Learning and Memory Deficit in Mice. Sci Rep 2016; 6:21186. [PMID: 26879001 PMCID: PMC4754754 DOI: 10.1038/srep21186] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/19/2016] [Indexed: 01/13/2023] Open
Abstract
Elderly individuals are at increased risk of cognitive decline after anesthesia. General anesthesia is believed to be a risk factor for Alzheimer’s disease (AD). At present, there is no treatment that can prevent anesthesia-induced postoperative cognitive dysfunction. Here, we treated mice with daily intranasal administration of insulin (1.75 U/day) for one week before anesthesia induced by intraperitoneal injection of propofol and maintained by inhalation of sevoflurane for 1 hr. We found that the insulin treatment prevented anesthesia-induced deficit in spatial learning and memory, as measured by Morris water maze task during 1–5 days after exposure to anesthesia. The insulin treatment also attenuated anesthesia-induced hyperphosphorylation of tau and promoted the expression of synaptic proteins and insulin signaling in the brain. These findings show a therapeutic potential of intranasal administration of insulin before surgery to reduce the risk of anesthesia-induced cognitive decline and AD.
Collapse
|
72
|
Wang C, Zhang F, Jiang S, Siedlak SL, Shen L, Perry G, Wang X, Tang B, Zhu X. Estrogen receptor-α is localized to neurofibrillary tangles in Alzheimer's disease. Sci Rep 2016; 6:20352. [PMID: 26837465 PMCID: PMC4738266 DOI: 10.1038/srep20352] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/30/2015] [Indexed: 01/30/2023] Open
Abstract
The female predominance for developing Alzheimer disease (AD) suggests the involvement of gender specific factor(s) such as a reduced estrogen-estrogen receptor signaling in the pathogenesis of AD. The potential role of ERα in AD pathogenesis has been explored by several groups with mixed results. We revisited this issue of expression and distribution of ERα in AD brain using a specific ERα antibody. Interestingly, we found that ERα co-localized with neurofibrillary pathology in AD brain and further demonstrated that ERα interacts with tau protein in vivo. Immunoprecipitaion experiments found increased ERα-tau interaction in the AD cases, which may account for ERα being sequestered in neuronal tau pathology. Indeed, tau overexpression in M17 cells leads to interruption of estrogen signaling. Our data support the idea that sequestration of ERα by tau pathology underlies the loss of estrogen neuroprotection during the course of AD.
Collapse
Affiliation(s)
- Chunyu Wang
- Department of Neurology, the second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fan Zhang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurosurgery, Chengdu first people’s Hospital, Chengdu, The People’s Republic of China
| | - Sirui Jiang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sandra L. Siedlak
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - George Perry
- Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
73
|
Day RJ, Mason MJ, Thomas C, Poon WW, Rohn TT. Caspase-Cleaved Tau Co-Localizes with Early Tangle Markers in the Human Vascular Dementia Brain. PLoS One 2015; 10:e0132637. [PMID: 26161867 PMCID: PMC4498690 DOI: 10.1371/journal.pone.0132637] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 06/16/2015] [Indexed: 02/01/2023] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia in the United States and is characterized as a cerebral vessel vascular disease that leads to ischemic episodes. Whereas the relationship between caspase-cleaved tau and neurofibrillary tangles (NFTs) in Alzheimer's disease (AD) has been previously described, whether caspase activation and cleavage of tau occurs in VaD is presently unknown. To investigate a potential role for caspase-cleaved tau in VaD, we analyzed seven confirmed cases of VaD by immunohistochemistry utilizing a well-characterized antibody that specifically detects caspase-cleaved tau truncated at Asp421. Application of this antibody (TauC3) revealed consistent labeling within NFTs, dystrophic neurites within plaque-rich regions and corpora amylacea (CA) in the human VaD brain. Labeling of CA by the TauC3 antibody was widespread throughout the hippocampus proper, was significantly higher compared to age matched controls, and co-localized with ubiquitin. Staining of the TauC3 antibody co-localized with MC-1, AT8, and PHF-1 within NFTs. Quantitative analysis indicated that roughly 90% of PHF-1-labeled NFTs contained caspase-cleaved tau. In addition, we documented the presence of active caspase-3 within plaques, blood vessels and pretangle neurons that co-localized with TauC3. Collectively, these data support a role for the activation of caspase-3 and proteolytic cleavage of TauC3 in VaD providing further support for the involvement of this family of proteases in NFT pathology.
Collapse
Affiliation(s)
- Ryan J. Day
- Department of Biological Sciences, Science Building, Room 228, Boise State University, Boise, Idaho, 83725, United States of America
| | - Maria J. Mason
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California, 92697, United States of America
| | - Chloe Thomas
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California, 92697, United States of America
| | - Wayne W. Poon
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California, 92697, United States of America
| | - Troy T. Rohn
- Department of Biological Sciences, Science Building, Room 228, Boise State University, Boise, Idaho, 83725, United States of America
- * E-mail:
| |
Collapse
|
74
|
Esteves-Villanueva JO, Trzeciakiewicz H, Martic S. A protein-based electrochemical biosensor for detection of tau protein, a neurodegenerative disease biomarker. Analyst 2015; 139:2823-31. [PMID: 24740472 DOI: 10.1039/c4an00204k] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A protein-based electrochemical biosensor was developed for detection of tau protein aimed towards electrochemically sensing misfolding proteins. The electrochemical assay monitors tau-tau binding and misfolding during the early stage of tau oligomerization. Electrochemical impedance spectroscopy was used to detect the binding event between solution tau protein and immobilized tau protein (tau-Au), acting as a recognition element. The charge transfer resistance (Rct) of tau-Au was 2.9 ± 0.6 kΩ. Subsequent tau binding to tau-Au decreased the Rct to 0.3 ± 0.1 kΩ (90 ± 3% decrease) upon formation of a tau-tau-Au interface. A linear relationship between the Rct and the solution tau concentration was observed from 0.2 to 1.0 μM. The Rct decrease was attributed to an enhanced charge permeability of the tau-tau-Au surface to a redox probe [Fe(CN)6](3-/4-). The electrochemical and surface characterization data suggested conformational and electrostatic changes induced by tau-tau binding. The protein-based electrochemical platform was highly selective for tau protein over bovine serum albumin and allowed for a rapid sample analysis. The protein-based interface was selective for a non-phosphorylated tau441 isoform over the paired-helical filaments of tau, which were composed of phosphorylated and truncated tau isoforms. The electrochemical approach may find application in screening of the early onset of neurodegeneration and aggregation inhibitors.
Collapse
|
75
|
Wheeler JM, McMillan PJ, Hawk M, Iba M, Robinson L, Xu GJ, Dombroski BA, Jeong D, Dichter MA, Juul H, Loomis E, Raskind M, Leverenz JB, Trojanowski JQ, Lee VM, Schellenberg GD, Kraemer BC. High copy wildtype human 1N4R tau expression promotes early pathological tauopathy accompanied by cognitive deficits without progressive neurofibrillary degeneration. Acta Neuropathol Commun 2015; 3:33. [PMID: 26041339 PMCID: PMC4453289 DOI: 10.1186/s40478-015-0210-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 05/04/2015] [Indexed: 12/31/2022] Open
Abstract
Introduction Accumulation of insoluble conformationally altered hyperphosphorylated tau occurs as part of the pathogenic process in Alzheimer’s disease (AD) and other tauopathies. In most AD subjects, wild-type (WT) tau aggregates and accumulates in neurofibrillary tangles and dystrophic neurites in the brain; however, in some familial tauopathy disorders, mutations in the gene encoding tau cause disease. Results We generated a mouse model, Tau4RTg2652, that expresses high levels of normal human tau in neurons resulting in the early stages of tau pathology. In this model, over expression of WT human tau drives pre-tangle pathology in young mice resulting in behavioral deficits. These changes occur at a relatively young age and recapitulate early pre-tangle stages of tau pathology associated with AD and mild cognitive impairment. Several features distinguish the Tau4RTg2652 model of tauopathy from previously described tau transgenic mice. Unlike other mouse models where behavioral and neuropathologic changes are induced by transgenic tau harboring MAPT mutations pathogenic for frontotemporal lobar degeneration (FTLD), the mice described here express the normal tau sequence. Conclusions Features of Tau4RTg2652 mice distinguishing them from other established wild type tau overexpressing mice include very early phenotypic manifestations, non-progressive tau pathology, abundant pre-tangle and phosphorylated tau, sparse oligomeric tau species, undetectable fibrillar tau pathology, stability of tau transgene copy number/expression, and normal lifespan. These results suggest that Tau4RTg2652 animals may facilitate studies of tauopathy target engagement where WT tau is driving tauopathy phenotypes. Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0210-6) contains supplementary material, which is available to authorized users.
Collapse
|
76
|
Plum S, Steinbach S, Abel L, Marcus K, Helling S, May C. Proteomics in neurodegenerative diseases: Methods for obtaining a closer look at the neuronal proteome. Proteomics Clin Appl 2014; 9:848-71. [DOI: 10.1002/prca.201400030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/25/2014] [Accepted: 09/03/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Sarah Plum
- Medizinisches Proteom-Center; Funktionelle Proteomik; Ruhr-Universität Bochum; Bochum Germany
| | - Simone Steinbach
- Medizinisches Proteom-Center; Medical Proteomics/Bioanalytics; Ruhr-Universität Bochum; Bochum Germany
| | - Laura Abel
- Medizinisches Proteom-Center; Medical Proteomics/Bioanalytics; Ruhr-Universität Bochum; Bochum Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center; Funktionelle Proteomik; Ruhr-Universität Bochum; Bochum Germany
| | - Stefan Helling
- Medizinisches Proteom-Center; Funktionelle Proteomik; Ruhr-Universität Bochum; Bochum Germany
| | - Caroline May
- Medizinisches Proteom-Center; Medical Proteomics/Bioanalytics; Ruhr-Universität Bochum; Bochum Germany
| |
Collapse
|
77
|
Frequent and symmetric deposition of misfolded tau oligomers within presynaptic and postsynaptic terminals in Alzheimer's disease. Acta Neuropathol Commun 2014; 2:146. [PMID: 25330988 PMCID: PMC4209049 DOI: 10.1186/s40478-014-0146-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 07/25/2014] [Indexed: 11/10/2022] Open
Abstract
The accumulation of neurofibrillary tangles in Alzheimer’s disease (AD) propagates with characteristic spatiotemporal patterns which follow brain network connections, implying trans-synaptic transmission of tauopathy. Since misfolded tau has been shown to transmit across synapses in AD animal models, we hypothesized that synapses in AD patients may contain misfolded tau. By immunofluorescence imaging of bipartite synapses from AD subjects, we detected tau protein in 38.4% of presynaptic and 50.9% of postsynaptic terminals. The pre/post distribution for hyperphosphorylated tau was 26.9%/30.7%, and for misfolded tau 18.3%/19.3%. In the temporal cortex, microscopic aggregates of tau, containing ultra-stable oligomers, were estimated to accumulate within trillions of synapses, outnumbering macroscopic tau aggregates such as tangles by 10000 fold. Non-demented elderly also showed considerable synaptic tau hyperphosphorylation and some misfolding, implicating the synapse as one of the first subcellular compartments affected by tauopathy. Misfolding of tau protein appeared to occur in situ inside synaptic terminals, without mislocalizing or mistrafficking. Misfolded tau at synapses may represent early signs of neuronal degeneration, mediators of synaptotoxicity, and anatomical substrates for transmitting tauopathy, but its actual role in these processes remain to be elucidated.
Collapse
|
78
|
Sontag JM, Wasek B, Taleski G, Smith J, Arning E, Sontag E, Bottiglieri T. Altered protein phosphatase 2A methylation and Tau phosphorylation in the young and aged brain of methylenetetrahydrofolate reductase (MTHFR) deficient mice. Front Aging Neurosci 2014; 6:214. [PMID: 25202269 PMCID: PMC4141544 DOI: 10.3389/fnagi.2014.00214] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 08/04/2014] [Indexed: 11/13/2022] Open
Abstract
Common functional polymorphisms in the methylenetetrahydrofolate reductase (MTHFR) gene, a key enzyme in folate and homocysteine metabolism, influence risk for a variety of complex disorders, including developmental, vascular, and neurological diseases. MTHFR deficiency is associated with elevation of homocysteine levels and alterations in the methylation cycle. Here, using young and aged Mthfr knockout mouse models, we show that mild MTHFR deficiency can lead to brain-region specific impairment of the methylation of Ser/Thr protein phosphatase 2A (PP2A). Relative to wild-type controls, decreased expression levels of PP2A and leucine carboxyl methyltransferase (LCMT1) were primarily observed in the hippocampus and cerebellum, and to a lesser extent in the cortex of young null Mthfr (-/-) and aged heterozygous Mthfr (+/-) mice. A marked down regulation of LCMT1 correlated with the loss of PP2A/Bα holoenzymes. Dietary folate deficiency significantly decreased LCMT1, methylated PP2A and PP2A/Bα levels in all brain regions examined from aged Mthfr (+/+) mice, and further exacerbated the regional effects of MTHFR deficiency in aged Mthfr (+/-) mice. In turn, the down regulation of PP2A/Bα was associated with enhanced phosphorylation of Tau, a neuropathological hallmark of Alzheimer's disease (AD). Our findings identify hypomethylation of PP2A enzymes, which are major CNS phosphatases, as a novel mechanism by which MTHFR deficiency and Mthfr gene-diet interactions could lead to disruption of neuronal homeostasis, and increase the risk for a variety of neuropsychiatric disorders, including age-related diseases like sporadic AD.
Collapse
Affiliation(s)
- Jean-Marie Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute Callaghan, NSW, Australia
| | - Brandi Wasek
- Institute of Metabolic Disease and Baylor Research Institute, Baylor University Medical Center Dallas, TX, USA
| | - Goce Taleski
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute Callaghan, NSW, Australia
| | - Josephine Smith
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute Callaghan, NSW, Australia
| | - Erland Arning
- Institute of Metabolic Disease and Baylor Research Institute, Baylor University Medical Center Dallas, TX, USA
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute Callaghan, NSW, Australia
| | - Teodoro Bottiglieri
- Institute of Metabolic Disease and Baylor Research Institute, Baylor University Medical Center Dallas, TX, USA
| |
Collapse
|
79
|
Ittner A, Bertz J, Suh LS, Stevens CH, Götz J, Ittner LM. Tau-targeting passive immunization modulates aspects of pathology in tau transgenic mice. J Neurochem 2014; 132:135-45. [DOI: 10.1111/jnc.12821] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/29/2014] [Accepted: 06/30/2014] [Indexed: 12/28/2022]
Affiliation(s)
- Arne Ittner
- Dementia Research Unit; School of Medical Sciences; The University of New South Wales; Sydney NSW Australia
- Brain & Mind Research Institute; The University of Sydney; Sydney NSW Australia
| | - Josefine Bertz
- Dementia Research Unit; School of Medical Sciences; The University of New South Wales; Sydney NSW Australia
- Brain & Mind Research Institute; The University of Sydney; Sydney NSW Australia
| | - Lisa S. Suh
- Dementia Research Unit; School of Medical Sciences; The University of New South Wales; Sydney NSW Australia
| | - Claire H. Stevens
- Dementia Research Unit; School of Medical Sciences; The University of New South Wales; Sydney NSW Australia
| | - Jürgen Götz
- Brain & Mind Research Institute; The University of Sydney; Sydney NSW Australia
- Clem Jones Centre for Ageing Dementia Research; Queensland Brain Institute; University of Queensland; Brisbane Qld. Australia
| | - Lars M. Ittner
- Dementia Research Unit; School of Medical Sciences; The University of New South Wales; Sydney NSW Australia
- Brain & Mind Research Institute; The University of Sydney; Sydney NSW Australia
- Neuroscience Research Australia; Sydney NSW Australia
| |
Collapse
|
80
|
Arnold SE, Toledo JB, Appleby DH, Xie SX, Wang LS, Baek Y, Wolk DA, Lee EB, Miller BL, Lee VMY, Trojanowski JQ. Comparative survey of the topographical distribution of signature molecular lesions in major neurodegenerative diseases. J Comp Neurol 2014; 521:4339-55. [PMID: 23881776 DOI: 10.1002/cne.23430] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/07/2013] [Accepted: 07/10/2013] [Indexed: 11/06/2022]
Abstract
An understanding of the anatomic distributions of major neurodegenerative disease lesions is important to appreciate the differential clinical profiles of these disorders and to serve as neuropathological standards for emerging molecular neuroimaging methods. To address these issues, here we present a comparative survey of the topographical distribution of the defining molecular neuropathological lesions among 10 neurodegenerative diseases from a large and uniformly assessed brain collection. Ratings of pathological severity in 16 brain regions from 671 cases with diverse neurodegenerative diseases are summarized and analyzed. These include: 1) amyloid-β and tau lesions in Alzheimer's disease; 2) tau lesions in three other tauopathies including Pick's disease, progressive supranuclear palsy and corticobasal degeneration; 3) α-synuclein inclusion ratings in four synucleinopathies including Parkinson's disease, Parkinson's disease with dementia, dementia with Lewy bodies, and multiple system atrophy; and 4) TDP-43 lesions in two TDP-43 proteinopathies, including frontotemporal lobar degeneration associated with TDP-43 and amyotrophic lateral sclerosis. The data presented graphically and topographically confirm and extend previous pathological anatomic descriptions and statistical comparisons highlight the lesion distributions that either overlap or distinguish the diseases in each molecular disease category.
Collapse
Affiliation(s)
- Steven E Arnold
- Penn Memory Center, Departments of Psychiatry and Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, 19104
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Di Meco A, Joshi YB, Praticò D. Sleep deprivation impairs memory, tau metabolism, and synaptic integrity of a mouse model of Alzheimer's disease with plaques and tangles. Neurobiol Aging 2014; 35:1813-20. [PMID: 24629673 DOI: 10.1016/j.neurobiolaging.2014.02.011] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/06/2014] [Accepted: 02/10/2014] [Indexed: 11/17/2022]
Abstract
Several studies have highlighted the frequency of sleep disturbances in Alzheimer's disease (AD). However, whether they are secondary to the disease or per se increase its risk remains to be fully investigated. The aim of the current investigation was to study the effect of sleep deprivation (SD) on the development of AD phenotype in a transgenic mouse model with plaques and tangles, the 3xTg mice. We evaluated the functional and biological consequences on 3xTg mice that underwent 4 hours sleep restrain per day for 8 weeks. Compared with controls, behavioral assessment showed that SD-treated mice had a significant decline in their learning and memory. Although no differences were detected in the levels of soluble amyloid-β peptides, the same animals displayed a decrease in tau phosphorylation, which associated with a significant increase in its insoluble fraction. In addition, we observed that SD resulted in lower levels of postsynaptic density protein 95 and increased glial fibrillary acidic protein levels. Finally, although total levels of the transcription factor cellular response element binding protein were unchanged, its phosphorylated form was significantly diminished in brains of sleep-deprived mice when compared with controls. Our study underlines the importance of SD as a chronic stressor, which by modulating biochemical processes influences the development of memory impairments and AD neuropathologies. Correction of SD could be a viable therapeutic strategy to prevent the onset or slow the progression of AD in individuals bearing this risk factor.
Collapse
Affiliation(s)
- Antonio Di Meco
- Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Yash B Joshi
- Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Domenico Praticò
- Department of Pharmacology, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
82
|
Soluble pathological tau in the entorhinal cortex leads to presynaptic deficits in an early Alzheimer's disease model. Acta Neuropathol 2014; 127:257-70. [PMID: 24271788 DOI: 10.1007/s00401-013-1215-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 10/28/2013] [Accepted: 11/09/2013] [Indexed: 10/26/2022]
Abstract
Neurofibrillary tangles (NFTs), a hallmark of Alzheimer's disease, are intracellular silver and thioflavin S-staining aggregates that emerge from earlier accumulation of phospho-tau in the soma. Whether soluble misfolded but nonfibrillar tau disrupts neuronal function is unclear. Here we investigate if soluble pathological tau, specifically directed to the entorhinal cortex (EC), can cause behavioral or synaptic deficits. We studied rTgTauEC transgenic mice, in which P301L mutant human tau overexpressed primarily in the EC leads to the development of tau pathology, but only rare NFT at 16 months of age. We show that the early tau lesions are associated with nearly normal performance in contextual fear conditioning, a hippocampal-related behavior task, but more robust changes in neuronal system activation as marked by Arc induction and clear electrophysiological defects in perforant pathway synaptic plasticity. Electrophysiological changes were likely due to a presynaptic deficit and changes in probability of neurotransmitter release. The data presented here support the hypothesis that misfolded and hyperphosphorylated tau can impair neuronal function within the entorhinal-hippocampal network, even prior to frank NFT formation and overt neurodegeneration.
Collapse
|
83
|
Siman R, Lin YG, Malthankar-Phatak G, Dong Y. A rapid gene delivery-based mouse model for early-stage Alzheimer disease-type tauopathy. J Neuropathol Exp Neurol 2013; 72:1062-71. [PMID: 24128676 PMCID: PMC3815088 DOI: 10.1097/nen.0000000000000006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The perforant pathway projection from the entorhinal cortex (EC) to the hippocampal dentate gyrus is critically important for long-term memory and develops tau and amyloid pathologies and progressive degeneration starting in the early stages of Alzheimer disease (AD). However, perforant pathway function has not been assessed in experimental models of AD, and a therapeutic agent that protects its structure and function has not yet been identified. Therefore, we developed a new adeno-associated virus-based mouse model for perforant pathway tauopathy. Microinjection into the lateral EC of vectors designed to express either human tau bearing a pathogenic P301L mutation or enhanced green fluorescent protein as a control selectively drove transgene expression in lateral EC layer II perikarya and along the entire rostrocaudal extent of the lateral perforant pathway afferents and dentate terminal field. After human tau expression, hyperphosphorylated tau accumulated only within EC layer II perikarya, thereby modeling Braak stage I of transentorhinal AD tauopathy. Expression of pathologic human tau but not enhanced green fluorescent protein led to specific dose-dependent apoptotic death of perforant pathway neurons and loss of synapses in as little as 2 weeks. This novel adeno-associated virus-based method elicits rapid tauopathy and tau-mediated neurodegeneration localized to the mouse perforant pathway and represents a new experimental approach for studying tau-driven pathogenic processes and tau-based treatment strategies in a highly vulnerable neural circuit.
Collapse
Affiliation(s)
- Robert Siman
- From the Department of Neurosurgery and Center for Brain Injury and Repair, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|
84
|
Kopeikina KJ, Polydoro M, Tai HC, Yaeger E, Carlson GA, Pitstick R, Hyman BT, Spires-Jones TL. Synaptic alterations in the rTg4510 mouse model of tauopathy. J Comp Neurol 2013; 521:1334-53. [PMID: 23047530 DOI: 10.1002/cne.23234] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/07/2012] [Accepted: 10/02/2012] [Indexed: 01/01/2023]
Abstract
Synapse loss, rather than the hallmark amyloid-β (Aβ) plaques or tau-filled neurofibrillary tangles (NFT), is considered the most predictive pathological feature associated with cognitive status in the Alzheimer's disease (AD) brain. The role of Aβ in synapse loss is well established, but despite data linking tau to synaptic function, the role of tau in synapse loss remains largely undetermined. Here we test the hypothesis that human mutant P301L tau overexpression in a mouse model (rTg4510) will lead to age-dependent synaptic loss and dysfunction. Using array tomography and two methods of quantification (automated, threshold-based counting and a manual stereology-based technique) we demonstrate that overall synapse density is maintained in the neuropil, implicating synapse loss commensurate with the cortical atrophy known to occur in this model. Multiphoton in vivo imaging reveals close to 30% loss of apical dendritic spines of individual pyramidal neurons, suggesting these cells may be particularly vulnerable to tau-induced degeneration. Postmortem, we confirm the presence of tau in dendritic spines of rTg4510-YFP mouse brain by array tomography. These data implicate tau-induced loss of a subset of synapses that may be accompanied by compensatory increases in other synaptic subtypes, thereby preserving overall synapse density. Biochemical fractionation of synaptosomes from rTg4510 brain demonstrates a significant decrease in expression of several synaptic proteins, suggesting a functional deficit of remaining synapses in the rTg4510 brain. Together, these data show morphological and biochemical synaptic consequences in response to tau overexpression in the rTg4510 mouse model.
Collapse
Affiliation(s)
- Katherine J Kopeikina
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
85
|
R-flurbiprofen improves tau, but not Aß pathology in a triple transgenic model of Alzheimer's disease. Brain Res 2013; 1541:115-27. [PMID: 24161403 DOI: 10.1016/j.brainres.2013.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 10/10/2013] [Accepted: 10/15/2013] [Indexed: 11/24/2022]
Abstract
We have previously reported that chronic ibuprofen treatment improves cognition and decreases intracellular Aß and phosphorylated-tau levels in 3xTg-AD mice. Ibuprofen is a non-steroidal anti-inflammatory drug (NSAID) that independently of its anti-inflammatory effects has anti-amyloidogenic activity as a gamma-secretase modulator (GSM) and both activities have the potential to decrease Aß pathology. To further understand the effects of NSAIDs in 3xTg-AD mice, we treated 3xTg-AD mice with R-flurbiprofen, an enantiomer of the NSAID flurbiprofen that maintains the GSM activity but has greatly reduced anti-inflammatory activity, and analyzed its effect on cognition, Aß, tau, and the neurochemical profile of the hippocampus. Treatment with R-flurbiprofen from 5 to 7 months of age resulted in improved cognition on the radial arm water maze (RAWM) test and decreased the level of hyperphosphorylated tau immunostained with AT8 and PHF-1 antibodies. No significant changes in the level of Aß (using 6E10 and NU-1 antibodies) were detected. Using magnetic resonance spectroscopy (MRS) we found that R-flurbiprofen treatment decreased the elevated level of glutamine in 3xTg-AD mice down to the level detected in non-transgenic mice. Glutamine levels correlated with PHF-1 immunostained hyperphosphorylated tau. We also found an inverse correlation between the concentration of glutamate and learning across all the mice in the study. Glutamine and glutamate, neurochemicals that shuttles between neurons and astrocytes to maintain glutamate homeostasis in the synapses, deserve further attention as MR markers of cognitive function.
Collapse
|
86
|
Affiliation(s)
- Brian H. Anderton
- Department of Neuroscience, Institute of Psychiatry, University of London, London, U.K
| |
Collapse
|
87
|
Zhang X, Hernandez I, Rei D, Mair W, Laha JK, Cornwell ME, Cuny GD, Tsai LH, Steen JAJ, Kosik KS. Diaminothiazoles modify Tau phosphorylation and improve the tauopathy in mouse models. J Biol Chem 2013; 288:22042-56. [PMID: 23737518 PMCID: PMC3724657 DOI: 10.1074/jbc.m112.436402] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 05/31/2013] [Indexed: 01/01/2023] Open
Abstract
Although Tau accumulation is a feature of several neurodegenerative conditions, treatment options for these conditions are nonexistent. Targeting Tau kinases represents a potential therapeutic approach. Small molecules in the diaminothiazole class are potent Tau kinase inhibitors that target CDK5 and GSK3β. Lead compounds from the series have IC50 values toward CDK5/p25 and GSK3β in the low nanomolar range and no observed toxicity in the therapeutic dose range. Neuronal protective effects and decreased PHF-1 immunoreactivity were observed in two animal models, 3×Tg-AD and CK-p25. Treatment nearly eliminated Sarkosyl-insoluble Tau with the most prominent effect on the phosphorylation at Ser-404. Treatment also induced the recovery of memory in a fear conditioning assay. Given the contribution of both CDK5/p25 and GSK3β to Tau phosphorylation, effective treatment of tauopathies may require dual kinase targeting.
Collapse
Affiliation(s)
- Xuemei Zhang
- From the Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California 93106
| | - Israel Hernandez
- From the Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California 93106
| | - Damien Rei
- the Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Waltraud Mair
- the F. M. Kirby Neurobiology Center, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts 02115
| | - Joydev K. Laha
- the Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts 02139
| | - Madison E. Cornwell
- From the Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California 93106
| | - Gregory D. Cuny
- the Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts 02139
| | - Li-Huei Tsai
- the Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Judith A. J. Steen
- the F. M. Kirby Neurobiology Center, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts 02115
| | - Kenneth S. Kosik
- From the Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California 93106
| |
Collapse
|
88
|
Abstract
The pathway leading from soluble and monomeric to hyperphosphorylated, insoluble and filamentous tau protein is at the centre of many human neurodegenerative diseases, collectively referred to as tauopathies. Dominantly inherited mutations in MAPT, the gene that encodes tau, cause forms of frontotemporal dementia and parkinsonism, proving that dysfunction of tau is sufficient to cause neurodegeneration and dementia. However, most cases of tauopathy are not inherited in a dominant manner. The first tau aggregates form in a few nerve cells in discrete brain areas. These become self propagating and spread to distant brain regions in a prion-like manner. The prevention of tau aggregation and propagation is the focus of attempts to develop mechanism-based treatments for tauopathies.
Collapse
Affiliation(s)
- Maria Grazia Spillantini
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
89
|
Rasool S, Martinez-Coria H, Wu JW, LaFerla F, Glabe CG. Systemic vaccination with anti-oligomeric monoclonal antibodies improves cognitive function by reducing Aβ deposition and tau pathology in 3xTg-AD mice. J Neurochem 2013; 126:473-82. [PMID: 23672786 DOI: 10.1111/jnc.12305] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/23/2013] [Accepted: 04/30/2013] [Indexed: 02/03/2023]
Abstract
Alzheimer's disease (AD) is a devastating disorder that is clinically characterized by a comprehensive cognitive decline. Accumulation of the amyloid-beta (Aβ) peptide plays a pivotal role in the pathogenesis of AD. In AD, the conversion of Aβ from a physiological soluble monomeric form into insoluble fibrillar conformation is an important event. The most toxic form of Aβ is oligomers, which is the intermediate step during the conversion of monomeric form to fibrillar form. There are at least two types of oligomers: oligomers that are immunologically related to fibrils and those that are not. In transgenic AD animal models, both active and passive anti-Aβ immunotherapies improve cognitive function and clear the parenchymal accumulation of amyloid plaques in the brain. In this report we studied effect of immunotherapy of two sequence-independent non-fibrillar oligomer specific monoclonal antibodies on the cognitive function, amyloid load and tau pathology in 3xTg-AD mice. Anti-oligomeric monoclonal antibodies significantly reduce the amyloid load and improve the cognition. The clearance of amyloid load was significantly correlated with reduced tau hyperphosphorylation and improvement in cognition. These results demonstrate that systemic immunotherapy using oligomer-specific monoclonal antibodies effectively attenuates behavioral and pathological impairments in 3xTg-AD mice. These findings demonstrate the potential of using oligomer specific monoclonal antibodies as a therapeutic approach to prevent and treat Alzheimer's disease.
Collapse
Affiliation(s)
- Suhail Rasool
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA.
| | | | | | | | | |
Collapse
|
90
|
Rasool S, Martinez-Coria H, Milton S, Glabe CG. Nonhuman amyloid oligomer epitope reduces Alzheimer's-like neuropathology in 3xTg-AD transgenic mice. Mol Neurobiol 2013; 48:931-40. [PMID: 23771815 DOI: 10.1007/s12035-013-8478-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 05/30/2013] [Indexed: 12/22/2022]
Abstract
Accumulation of beta-amyloid (Aβ) is an important pathological event in Alzheimer's disease (AD). It is now well known that vaccination against fibrillar Aβ prevents amyloid accumulation and preserves cognitive function in transgenic mouse models. To study the effect of vaccination against generic oligomer epitopes, Aβ oligomers, islet amyloid polypeptide oligomers, random peptide oligomer (3A), and Aβ fibrils were used to vaccinate 3xTg-AD, which develop a progressive accumulation of plaques and cognitive impairment. Subcutaneous administration of these antigens markedly reduced total plaque load (Aβ burden) and improved cognitive function in the 3xTg-AD mouse brains as compared to controls. We demonstrated that vaccination with this nonhuman amyloid oligomer generated high titers of specifically antibodies recognizing Aβ oligomers, which in turn inhibited accumulation of Aβ pathology in mice. In addition to amyloid plaques, another hallmark of AD is tau pathology. It was found that there was a significant decline in the level of hyper-phosphorylated tau following vaccination. We have previously shown that immunization with 3A peptide improves cognitive function and clears amyloid plaques in Tg2576 mice, which provides a novel strategy of AD therapy. Here, we have shown that vaccination with 3A peptide in 3xTg-AD mice not only clears amyloid plaques but also extensively clears abnormal tau in brain.
Collapse
Affiliation(s)
- Suhail Rasool
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, USA,
| | | | | | | |
Collapse
|
91
|
Clavaguera F, Lavenir I, Falcon B, Frank S, Goedert M, Tolnay M. "Prion-like" templated misfolding in tauopathies. Brain Pathol 2013; 23:342-9. [PMID: 23587140 PMCID: PMC8028860 DOI: 10.1111/bpa.12044] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 02/11/2013] [Indexed: 12/20/2022] Open
Abstract
The soluble microtubule-associated protein tau forms hyperphosphorylated, insoluble and filamentous inclusions in a number of neurodegenerative diseases referred to as "tauopathies." In Alzheimer's disease, tau pathology develops in a stereotypical manner, with the first lesions appearing in the locus coeruleus and entorhinal cortex, from where they appear to spread to the hippocampus and neocortex. Propagation of tau pathology is also a characteristic of argyrophilic grain disease, where the tau lesions spread throughout the limbic system. Significantly, isoform composition and morphology of tau filaments can differ between tauopathies, suggesting the existence of distinct tau strains. Extensive experimental findings indicate that prion-like mechanisms underly the pathogenesis of tauopathies.
Collapse
Affiliation(s)
- Florence Clavaguera
- Department of NeuropathologyInstitute of PathologyUniversity Hospital BaselBaselSwitzerland
| | | | - Ben Falcon
- MRC Laboratory of Molecular BiologyCambridgeUK
| | - Stephan Frank
- Department of NeuropathologyInstitute of PathologyUniversity Hospital BaselBaselSwitzerland
| | | | - Markus Tolnay
- Department of NeuropathologyInstitute of PathologyUniversity Hospital BaselBaselSwitzerland
| |
Collapse
|
92
|
Umeda T, Tomiyama T, Kitajima E, Idomoto T, Nomura S, Lambert MP, Klein WL, Mori H. Hypercholesterolemia accelerates intraneuronal accumulation of Aβ oligomers resulting in memory impairment in Alzheimer's disease model mice. Life Sci 2012; 91:1169-76. [DOI: 10.1016/j.lfs.2011.12.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 12/12/2011] [Accepted: 12/21/2011] [Indexed: 01/07/2023]
|
93
|
Tai HC, Serrano-Pozo A, Hashimoto T, Frosch MP, Spires-Jones TL, Hyman BT. The synaptic accumulation of hyperphosphorylated tau oligomers in Alzheimer disease is associated with dysfunction of the ubiquitin-proteasome system. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1426-35. [PMID: 22867711 DOI: 10.1016/j.ajpath.2012.06.033] [Citation(s) in RCA: 326] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/15/2012] [Accepted: 06/20/2012] [Indexed: 12/13/2022]
Abstract
In Alzheimer disease (AD), deposition of neurofibrillary tangles and loss of synapses in the neocortex and limbic system each correlate strongly with cognitive impairment. Tangles are composed of misfolded hyperphosphorylated tau proteins; however, the link between tau abnormalities and synaptic dysfunction remains unclear. We examined the location of tau in control and AD cortices using biochemical and morphologic methods. We found that, in addition to its well-described axonal localization, normal tau is present at both presynaptic and postsynaptic terminals in control human brains. In AD, tau becomes hyperphosphorylated and misfolded at both presynaptic and postsynaptic terminals, and this abnormally posttranslationally modified tau is enriched in synaptoneurosomal fractions. Synaptic tau seems to be hyperphosphorylated and ubiquitinated, and forms stable oligomers resistant to SDS denaturation. The accumulation of hyperphosphorylated tau oligomers at human AD synapses is associated with increased ubiquitinated substrates and increased proteasome components, consistent with dysfunction of the ubiquitin-proteasome system. Our findings suggest that synaptic hyperphosphorylated tau oligomers may be an important mediator of the proteotoxicity that disrupts synapses in AD.
Collapse
Affiliation(s)
- Hwan-Ching Tai
- MassGeneral Institute of Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
94
|
Acker CM, Forest SK, Zinkowski R, Davies P, d'Abramo C. Sensitive quantitative assays for tau and phospho-tau in transgenic mouse models. Neurobiol Aging 2012; 34:338-50. [PMID: 22727277 DOI: 10.1016/j.neurobiolaging.2012.05.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/16/2012] [Accepted: 05/21/2012] [Indexed: 12/20/2022]
Abstract
Transgenic mouse models have been an invaluable resource in elucidating the complex roles of β-amyloid and tau in Alzheimer's disease. Although many laboratories rely on qualitative or semiquantitative techniques when investigating tau pathology, we have developed 4 Low-Tau, Sandwich enzyme-linked immunosorbent assays (ELISAs) that quantitatively assess different epitopes of tau relevant to Alzheimer's disease: total tau, pSer-202, pThr-231, and pSer-396/404. In this study, after comparing our assays with commercially available ELISAs, we demonstrate our assay's high specificity and quantitative capabilities using brain homogenates from tau transgenic mice, htau, JNPL3, and tau knockout. All 4 ELISAs show excellent specificity for mouse and human tau, with no reactivity to tau knockout animals. An age-dependent increase of serum tau in both tau transgenic models was also seen. Taken together, these assays are valuable methods to quantify tau and phospho-tau levels in transgenic animals, by examining tau levels in brain and measuring tau as a potential serum biomarker.
Collapse
Affiliation(s)
- Christopher M Acker
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | | | |
Collapse
|
95
|
Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, Greco A, Seripa D, Pilotto A. Immunotherapy for Alzheimer's disease: from anti-β-amyloid to tau-based immunization strategies. Immunotherapy 2012; 4:213-38. [PMID: 22339463 DOI: 10.2217/imt.11.170] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The exact mechanisms leading to Alzheimer's disease (AD) are largely unknown, limiting the identification of effective disease-modifying therapies. The two principal neuropathological hallmarks of AD are extracellular β-amyloid (Aβ), peptide deposition (senile plaques) and intracellular neurofibrillary tangles containing hyperphosphorylated tau protein. During the last decade, most of the efforts of the pharmaceutical industry were directed against the production and accumulation of Aβ. The most innovative of the pharmacological approaches was the stimulation of Aβ clearance from the brain of AD patients via the administration of Aβ antigens (active vaccination) or anti-Aβ antibodies (passive vaccination). Several active and passive anti-Aβ vaccines are under clinical investigation. Unfortunately, the first active vaccine (AN1792, consisting of preaggregate Aβ and an immune adjuvant, QS-21) was abandoned because it caused meningoencephalitis in approximately 6% of treated patients. Anti-Aβ monoclonal antibodies (bapineuzumab and solanezumab) are now being developed. The clinical results of the initial studies with bapineuzumab were equivocal in terms of cognitive benefit. The occurrence of vasogenic edema after bapineuzumab, and more rarely brain microhemorrhages (especially in Apo E ε4 carriers), has raised concerns on the safety of these antibodies directed against the N-terminus of the Aβ peptide. Solanezumab, a humanized anti-Aβ monoclonal antibody directed against the midregion of the Aβ peptide, was shown to neutralize soluble Aβ species. Phase II studies showed a good safety profile of solanezumab, while studies on cerebrospinal and plasma biomarkers documented good signals of pharmacodynamic activity. Although some studies suggested that active immunization may be effective against tau in animal models of AD, very few studies regarding passive immunization against tau protein are currently available. The results of the large, ongoing Phase III trials with bapineuzumab and solanezumab will tell us if monoclonal anti-Aβ antibodies may slow down the rate of deterioration of AD. Based on the new diagnostic criteria of AD and on recent major failures of anti-Aβ drugs in mild-to-moderate AD patients, one could argue that clinical trials on potential disease-modifying drugs, including immunological approaches, should be performed in the early stages of AD.
Collapse
Affiliation(s)
- Francesco Panza
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, Foggia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Álvarez G, Aldudo J, Alonso M, Santana S, Valdivieso F. Herpes simplex virus type 1 induces nuclear accumulation of hyperphosphorylated tau in neuronal cells. J Neurosci Res 2012; 90:1020-9. [DOI: 10.1002/jnr.23003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 11/04/2011] [Accepted: 11/12/2011] [Indexed: 12/20/2022]
|
97
|
Valvezan AJ, Zhang F, Diehl JA, Klein PS. Adenomatous polyposis coli (APC) regulates multiple signaling pathways by enhancing glycogen synthase kinase-3 (GSK-3) activity. J Biol Chem 2011; 287:3823-32. [PMID: 22184111 DOI: 10.1074/jbc.m111.323337] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is essential for many signaling pathways and cellular processes. As Adenomatous Polyposis Coli (APC) functions in many of the same processes, we investigated a role for APC in the regulation of GSK-3-dependent signaling. We find that APC directly enhances GSK-3 activity. Furthermore, knockdown of APC mimics inhibition of GSK-3 by reducing phosphorylation of glycogen synthase and by activating mTOR, revealing novel roles for APC in the regulation of these enzymes. Wnt signaling inhibits GSK-3 through an unknown mechanism, and this results in both stabilization of β-catenin and activation of mTOR. We therefore hypothesized that Wnts may regulate GSK-3 by disrupting the interaction between APC and the Axin-GSK-3 complex. We find that Wnts rapidly induce APC dissociation from Axin, correlating with β-catenin stabilization. Furthermore, Axin interaction with the Wnt co-receptor LRP6 causes APC dissociation from Axin. We propose that APC regulates multiple signaling pathways by enhancing GSK-3 activity, and that Wnts induce APC dissociation from Axin to reduce GSK-3 activity and activate downstream signaling. APC regulation of GSK-3 also provides a novel mechanism for Wnt regulation of multiple downstream effectors, including β-catenin and mTOR.
Collapse
Affiliation(s)
- Alexander J Valvezan
- Cell and Molecular Biology Graduate Group, The Leonard and Madlyn Abramson Family Cancer Research Institute and Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
98
|
Tran HT, Sanchez L, Esparza TJ, Brody DL. Distinct temporal and anatomical distributions of amyloid-β and tau abnormalities following controlled cortical impact in transgenic mice. PLoS One 2011; 6:e25475. [PMID: 21980472 PMCID: PMC3183029 DOI: 10.1371/journal.pone.0025475] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/05/2011] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) is a major environmental risk factor for Alzheimer's disease. Intracellular accumulations of amyloid-β and tau proteins have been observed within hours following severe TBI in humans. Similar abnormalities have been recapitulated in young 3xTg-AD mice subjected to the controlled cortical impact model (CCI) of TBI and sacrificed at 24 h and 7 days post injury. This study investigated the temporal and anatomical distributions of amyloid-β and tau abnormalities from 1 h to 24 h post injury in the same model. Intra-axonal amyloid-β accumulation in the fimbria was detected as early as 1 hour and increased monotonically over 24 hours following injury. Tau immunoreactivity in the fimbria and amygdala had a biphasic time course with peaks at 1 hour and 24 hours, while tau immunoreactivity in the contralateral CA1 rose in a delayed fashion starting at 12 hours after injury. Furthermore, rapid intra-axonal amyloid-β accumulation was similarly observed post controlled cortical injury in APP/PS1 mice, another transgenic Alzheimer's disease mouse model. Acute increases in total and phospho-tau immunoreactivity were also evident in single transgenic TauP301L mice subjected to controlled cortical injury. These data provide further evidence for the causal effects of moderately severe contusional TBI on acceleration of acute Alzheimer-related abnormalities and the independent relationship between amyloid-β and tau in this setting.
Collapse
Affiliation(s)
- Hien T. Tran
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Laura Sanchez
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Thomas J. Esparza
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - David L. Brody
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
99
|
Akundi RS, Zhi L, Büeler H. PINK1 enhances insulin-like growth factor-1-dependent Akt signaling and protection against apoptosis. Neurobiol Dis 2011; 45:469-78. [PMID: 21945539 DOI: 10.1016/j.nbd.2011.08.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/23/2011] [Accepted: 08/26/2011] [Indexed: 12/13/2022] Open
Abstract
Mutations in the PARK6 gene coding for PTEN-induced kinase 1 (PINK1) cause recessive early-onset Parkinsonism. Although PINK1 and Parkin promote the degradation of depolarized mitochondria in cultured cells, little is known about changes in signaling pathways that may additionally contribute to dopamine neuron loss in recessive Parkinsonism. Accumulating evidence implicates impaired Akt cell survival signaling in sporadic and familial PD (PD). IGF-1/Akt signaling inhibits dopamine neuron loss in several animal models of PD and both IGF-1 and insulin are neuroprotective in various settings. Here, we tested whether PINK1 is required for insulin-like growth factor 1 (IGF-1) and insulin dependent phosphorylation of Akt and the regulation of downstream Akt target proteins. Our results show that embryonic fibroblasts from PINK1-deficient mice display significantly reduced Akt phosphorylation in response to both IGF-1 and insulin. Moreover, phosphorylation of glycogen synthase kinase-3β (GSK-3β) and nuclear exclusion of FoxO1 are decreased in IGF-1 treated PINK1-deficient cells. In addition, phosphorylation of ribosomal protein S6 is reduced indicating decreased activity of mitochondrial target of rapamycin (mTOR) in IGF-1 treated PINK1(-/-) cells. Importantly, the protection afforded by IGF-1 against staurosporine-induced metabolic dysfunction and apoptosis is abrogated in PINK1-deficient cells. Moreover, IGF-1-induced Akt phosphorylation is impaired in primary cortical neurons from PINK1-deficient mice. Inhibition of cellular Ser/Thr phosphatases did not increase the amount of phosphorylated Akt in PINK1(-/-) cells, suggesting that components upstream of Akt phosphorylation are compromised in PINK1-deficient cells. Our studies show that PINK1 is required for optimal IGF-1 and insulin dependent Akt signal transduction, and raise the possibility that impaired IGF-1/Akt signaling is involved in PINK1-related Parkinsonism by increasing the vulnerability of dopaminergic neurons to stress-induced cell death.
Collapse
Affiliation(s)
- Ravi S Akundi
- Department of Anatomy and Neurobiology, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA
| | | | | |
Collapse
|
100
|
Soluble tau species, not neurofibrillary aggregates, disrupt neural system integration in a tau transgenic model. J Neuropathol Exp Neurol 2011; 70:588-95. [PMID: 21666499 DOI: 10.1097/nen.0b013e318220a658] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Neurofibrillary tangles are a feature of Alzheimer disease and other tauopathies, and although they are generally believed to be markers of neuronal pathology, there is little evidence evaluating whether tangles directly impact neuronal function. To investigate the response of cells in hippocampal circuits to complex behavioral stimuli, we used an environmental enrichment paradigm to induce expression of an immediate-early gene, Arc, in the rTg4510 mouse model of tauopathy. These mice reversibly overexpress P301L tau and exhibit substantial neurofibrillary tangle deposition, neuronal loss, and memory deficits. Using fluorescent in situ hybridization to detect Arc messenger RNA, we found that rTg4510 mice have impaired hippocampal Arc expression both without stimulation and in response to environmental enrichment; this likely reflects the combination of functional impairments of existing neurons and loss of neurons. However, tangle-bearing cells were at least as likely as non-tangle-bearing neurons to exhibit Arc expression in response to enrichment. Transgene suppression with doxycycline for 6 weeks resulted in increased percentages of Arc-positive cells in rTg4510 brains compared with untreated transgenics, restoring enrichment-induced Arc messenger RNA levels to that of wild-type controls despite the continued presence of neurofibrillary pathology. We interpret these data to indicate that soluble tau contributes to impairment of hippocampal function, although tangles do not preclude neurons from responding in a functional circuit.
Collapse
|