1
|
Goodman LD, Ralhan I, Li X, Lu S, Moulton MJ, Park YJ, Zhao P, Kanca O, Ghaderpour Taleghani ZS, Jacquemyn J, Shulman JM, Ando K, Sun K, Ioannou MS, Bellen HJ. Tau is required for glial lipid droplet formation and resistance to neuronal oxidative stress. Nat Neurosci 2024; 27:1918-1933. [PMID: 39187706 DOI: 10.1038/s41593-024-01740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 07/29/2024] [Indexed: 08/28/2024]
Abstract
The accumulation of reactive oxygen species (ROS) is a common feature of tauopathies, defined by Tau accumulations in neurons and glia. High ROS in neurons causes lipid production and the export of toxic peroxidated lipids (LPOs). Glia uptake these LPOs and incorporate them into lipid droplets (LDs) for storage and catabolism. We found that overexpressing Tau in glia disrupts LDs in flies and rat neuron-astrocyte co-cultures, sensitizing the glia to toxic, neuronal LPOs. Using a new fly tau loss-of-function allele and RNA-mediated interference, we found that endogenous Tau is required for glial LD formation and protection against neuronal LPOs. Similarly, endogenous Tau is required in rat astrocytes and human oligodendrocyte-like cells for LD formation and the breakdown of LPOs. Behaviorally, flies lacking glial Tau have decreased lifespans and motor defects that are rescuable by administering the antioxidant N-acetylcysteine amide. Overall, this work provides insights into the important role that Tau has in glia to mitigate ROS in the brain.
Collapse
Affiliation(s)
- Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Ye-Jin Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Program in Development, Disease Models and Therapeutics, Baylor College of Medicine, Houston, TX, USA
| | - Pinghan Zhao
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Ziyaneh S Ghaderpour Taleghani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Julie Jacquemyn
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Joshua M Shulman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Kanae Ando
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Program in Development, Disease Models and Therapeutics, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Zhang X, Wang J, Zhang Z, Ye K. Tau in neurodegenerative diseases: molecular mechanisms, biomarkers, and therapeutic strategies. Transl Neurodegener 2024; 13:40. [PMID: 39107835 PMCID: PMC11302116 DOI: 10.1186/s40035-024-00429-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/05/2024] [Indexed: 09/14/2024] Open
Abstract
The deposition of abnormal tau protein is characteristic of Alzheimer's disease (AD) and a class of neurodegenerative diseases called tauopathies. Physiologically, tau maintains an intrinsically disordered structure and plays diverse roles in neurons. Pathologically, tau undergoes abnormal post-translational modifications and forms oligomers or fibrous aggregates in tauopathies. In this review, we briefly introduce several tauopathies and discuss the mechanisms mediating tau aggregation and propagation. We also describe the toxicity of tau pathology. Finally, we explore the early diagnostic biomarkers and treatments targeting tau. Although some encouraging results have been achieved in animal experiments and preclinical studies, there is still no cure for tauopathies. More in-depth basic and clinical research on the pathogenesis of tauopathies is necessary.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiangyu Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Radosinska D, Radosinska J. The Link Between Matrix Metalloproteinases and Alzheimer's Disease Pathophysiology. Mol Neurobiol 2024:10.1007/s12035-024-04315-0. [PMID: 38935232 DOI: 10.1007/s12035-024-04315-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024]
Abstract
Alzheimer's disease (AD) is a major contributor to dementia and the most common neurodegenerative disorder. In AD pathophysiology, matrix metalloproteinases (MMPs)-proteolytic enzymes, best known to be responsible for remodeling and degradation of the extracellular matrix-were suggested to play an important role. Due to the diverse nature of the published data and frequent inconsistent results presented in available papers, it was considered essential to analyze all aspects of MMP literature with respect to AD pathophysiology and attempt to outline a unifying concept for understanding their role in AD. Thus, the main contribution of this review article is to summarize the most recent research on the participation of MMP in AD pathophysiology obtained using the cell cultures to understand the molecular principles of their action. Furthermore, an updated comprehensive view regarding this topic based exclusively on papers from human studies is provided as well. It can be concluded that determining the exact role of any particular MMPs in the AD pathophysiology holds promise for establishing their role as potential biomarkers reflecting the severity or progression of this disease or for developing new therapeutic agents targeting the processes that lead to AD.
Collapse
Affiliation(s)
- Dominika Radosinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Jana Radosinska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 81372, Bratislava, Slovak Republic.
| |
Collapse
|
4
|
Chinnathambi S, Chidambaram H. G-protein coupled receptors regulates Tauopathy in neurodegeneration. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:467-493. [PMID: 38960483 DOI: 10.1016/bs.apcsb.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
In Alzheimer's disease, the microtubule-associated protein, Tau misfolds to form aggregates and filaments in the intra- and extracellular region of neuronal cells. Microglial cells are the resident brain macrophage cells involved in constant surveillance and activated by the extracellular deposits. Purinergic receptors are involved in the chemotactic migration of microglial cells towards the site of inflammation. From our recent study, we have observed that the microglial P2Y12 receptor is involved in phagocytosis of full-length Tau species such as monomers, oligomers and aggregates by actin-driven chemotaxis. This study shows the interaction of repeat-domain of Tau (TauRD) with the microglial P2Y12 receptor and the corresponding residues for interaction have been analyzed by various in-silico approaches. In the cellular studies, TauRD was found to interact with microglial P2Y12R and induces its cellular expression confirmed by co-immunoprecipitation and western blot analysis. Furthermore, the P2Y12R-mediated TauRD internalization has demonstrated activation of microglia with an increase in the Iba1 level, and TauRD becomes accumulated at the peri-nuclear region for the degradation.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India.
| | - Hariharakrishnan Chidambaram
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| |
Collapse
|
5
|
Jamet M, Dupuis L, Gonzalez De Aguilar JL. Oligodendrocytes in amyotrophic lateral sclerosis and frontotemporal dementia: the new players on stage. Front Mol Neurosci 2024; 17:1375330. [PMID: 38585368 PMCID: PMC10995329 DOI: 10.3389/fnmol.2024.1375330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are fatal adult-onset neurodegenerative disorders that share clinical, neuropathological and genetic features, which forms part of a multi-system disease spectrum. The pathological process leading to ALS and FTD is the result of the combination of multiple mechanisms that operate within specific populations of neurons and glial cells. The implication of oligodendrocytes has been the subject of a number of studies conducted on patients and related animal models. In this review we summarize our current knowledge on the alterations specific to myelin and the oligodendrocyte lineage occurring in ALS and FTD. We also consider different ways by which specific oligodendroglial alterations influence neurodegeneration and highlight the important role of oligodendrocytes in these two intrinsically associated neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Jose-Luis Gonzalez De Aguilar
- Strasbourg Translational Neuroscience and Psychiatry, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
6
|
Congdon EE, Ji C, Tetlow AM, Jiang Y, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease: current status and future directions. Nat Rev Neurol 2023; 19:715-736. [PMID: 37875627 PMCID: PMC10965012 DOI: 10.1038/s41582-023-00883-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Alzheimer disease (AD) is the most common cause of dementia in older individuals. AD is characterized pathologically by amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the brain, with associated loss of synapses and neurons, which eventually results in dementia. Many of the early attempts to develop treatments for AD focused on Aβ, but a lack of efficacy of these treatments in terms of slowing disease progression led to a change of strategy towards targeting of tau pathology. Given that tau shows a stronger correlation with symptom severity than does Aβ, targeting of tau is more likely to be efficacious once cognitive decline begins. Anti-tau therapies initially focused on post-translational modifications, inhibition of tau aggregation and stabilization of microtubules. However, trials of many potential drugs were discontinued because of toxicity and/or lack of efficacy. Currently, the majority of tau-targeting agents in clinical trials are immunotherapies. In this Review, we provide an update on the results from the initial immunotherapy trials and an overview of new therapeutic candidates that are in clinical development, as well as considering future directions for tau-targeting therapies.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Changyi Ji
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Amber M Tetlow
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Brown RB. Dysregulated phosphate metabolism in autism spectrum disorder: associations and insights for future research. Expert Rev Mol Med 2023; 25:e20. [PMID: 37309057 PMCID: PMC10407224 DOI: 10.1017/erm.2023.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/27/2023] [Accepted: 05/09/2023] [Indexed: 06/14/2023]
Abstract
Studies of autism spectrum disorder (ASD) related to exposure to toxic levels of dietary phosphate are lacking. Phosphate toxicity from dysregulated phosphate metabolism can negatively impact almost every major organ system of the body, including the central nervous system. The present paper used a grounded theory-literature review method to synthesise associations of dysregulated phosphate metabolism with the aetiology of ASD. Cell signalling in autism has been linked to an altered balance between phosphoinositide kinases, which phosphorylate proteins, and the counteracting effect of phosphatases in neuronal membranes. Glial cell overgrowth in the developing ASD brain can lead to disturbances in neuro-circuitry, neuroinflammation and immune responses which are potentially related to excessive inorganic phosphate. The rise in ASD prevalence has been suggested to originate in changes to the gut microbiome from increasing consumption of additives in processed food, including phosphate additives. Ketogenic diets and dietary patterns that eliminate casein also reduce phosphate intake, which may account for many of the suggested benefits of these diets in children with ASD. Dysregulated phosphate metabolism is causatively linked to comorbid conditions associated with ASD such as cancer, tuberous sclerosis, mitochondrial dysfunction, diabetes, epilepsy, obesity, chronic kidney disease, tauopathy, cardiovascular disease and bone mineral disorders. Associations and proposals presented in this paper offer novel insights and directions for future research linking the aetiology of ASD with dysregulated phosphate metabolism and phosphate toxicity from excessive dietary phosphorus intake.
Collapse
Affiliation(s)
- Ronald B. Brown
- University of Waterloo, School of Public Health Sciences, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
8
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| |
Collapse
|
9
|
Miedema SSM, Mol MO, Koopmans FTW, Hondius DC, van Nierop P, Menden K, de Veij Mestdagh CF, van Rooij J, Ganz AB, Paliukhovich I, Melhem S, Li KW, Holstege H, Rizzu P, van Kesteren RE, van Swieten JC, Heutink P, Smit AB. Distinct cell type-specific protein signatures in GRN and MAPT genetic subtypes of frontotemporal dementia. Acta Neuropathol Commun 2022; 10:100. [PMID: 35799292 PMCID: PMC9261008 DOI: 10.1186/s40478-022-01387-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/22/2022] [Indexed: 11/16/2022] Open
Abstract
Frontotemporal dementia is characterized by progressive atrophy of frontal and/or temporal cortices at an early age of onset. The disorder shows considerable clinical, pathological, and genetic heterogeneity. Here we investigated the proteomic signatures of frontal and temporal cortex from brains with frontotemporal dementia due to GRN and MAPT mutations to identify the key cell types and molecular pathways in their pathophysiology. We compared patients with mutations in the GRN gene (n = 9) or with mutations in the MAPT gene (n = 13) with non-demented controls (n = 11). Using quantitative proteomic analysis on laser-dissected tissues we identified brain region-specific protein signatures for both genetic subtypes. Using published single cell RNA expression data resources we deduced the involvement of major brain cell types in driving these different protein signatures. Subsequent gene ontology analysis identified distinct genetic subtype- and cell type-specific biological processes. For the GRN subtype, we observed a distinct role for immune processes related to endothelial cells and for mitochondrial dysregulation in neurons. For the MAPT subtype, we observed distinct involvement of dysregulated RNA processing, oligodendrocyte dysfunction, and axonal impairments. Comparison with an in-house protein signature of Alzheimer’s disease brains indicated that the observed alterations in RNA processing and oligodendrocyte function are distinct for the frontotemporal dementia MAPT subtype. Taken together, our results indicate the involvement of different brain cell types and biological mechanisms in genetic subtypes of frontotemporal dementia. Furthermore, we demonstrate that comparison of proteomic profiles of different disease entities can separate general neurodegenerative processes from disease-specific pathways, which may aid the development of disease subtype-specific treatment strategies.
Collapse
Affiliation(s)
- Suzanne S M Miedema
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands.
| | - Merel O Mol
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frank T W Koopmans
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - David C Hondius
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Pim van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Kevin Menden
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany
| | - Christina F de Veij Mestdagh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands.,Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeroen van Rooij
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andrea B Ganz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands.,Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Iryna Paliukhovich
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Shamiram Melhem
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Henne Holstege
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Genetics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Impact of Partial Volume Correction on [18F]GE-180 PET Quantification in Subcortical Brain Regions of Patients with Corticobasal Syndrome. Brain Sci 2022; 12:brainsci12020204. [PMID: 35203967 PMCID: PMC8870519 DOI: 10.3390/brainsci12020204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
Corticobasal syndrome (CBS) is a rare neurodegenerative condition characterized by four-repeat tau aggregation in the cortical and subcortical brain regions and accompanied by severe atrophy. The aim of this study was to evaluate partial volume effect correction (PVEC) in patients with CBS compared to a control cohort imaged with the 18-kDa translocator protein (TSPO) positron emission tomography (PET) tracer [18F]GE-180. Eighteen patients with CBS and 12 age- and sex-matched healthy controls underwent [18F]GE-180 PET. The cortical and subcortical regions were delineated by deep nuclei parcellation (DNP) of a 3D-T1 MRI. Region-specific subcortical volumes and standardized uptake values and ratios (SUV and SUVr) were extracted before and after region-based voxel-wise PVEC. Regional volumes were compared between patients with CBS and controls. The % group differences and effect sizes (CBS vs. controls) of uncorrected and PVE-corrected SUVr data were compared. Single-region positivity in patients with CBS was assessed by a >2 SD threshold vs. controls and compared between uncorrected and PVE-corrected data. Smaller regional volumes were detected in patients with CBS compared to controls in the right ventral striatum (p = 0.041), the left putamen (p = 0.005), the right putamen (p = 0.038) and the left pallidum (p = 0.015). After applying PVEC, the % group differences were distinctly higher, but the effect sizes of TSPO uptake were only slightly stronger due to the higher variance after PVEC. The single-region positivity of TSPO PET increased in patients with CBS after PVEC (100 vs. 83 regions). PVEC in the cortical and subcortical regions is valuable for TSPO imaging of patients with CBS, leading to the improved detection of elevated [18F]GE-180 uptake, although the effect sizes in the comparison against the controls did not improve strongly.
Collapse
|
11
|
Vallés-Saiz L, Peinado-Cahuchola R, Ávila J, Hernández F. Microtubule-associated protein tau in murine kidney: role in podocyte architecture. Cell Mol Life Sci 2022; 79:97. [PMID: 35084555 PMCID: PMC8794918 DOI: 10.1007/s00018-021-04106-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/29/2021] [Accepted: 12/19/2021] [Indexed: 12/25/2022]
Abstract
Tau is a cytoskeletal protein that is expressed mainly in neurons and is involved in several cellular processes, such as microtubule stabilization, axonal maintenance, and transport. Altered tau metabolism is related to different tauopathies being the most important Alzheimer's disease where aberrant hyperphosphorylated and aggregated tau is found in the central nervous system. Here, we have analyzed that function in kidney by using tau knockout mice generated by integrating GFP-encoding cDNA into exon 1 of MAPT (here referred to as TauGFP/GFP). IVIS Lumina from PerkinElmer demonstrated GFP expression in the kidney. We then demonstrated by qPCR that the main tau isoform in the kidney is Tau4R. The GFP reporter allowed us to demonstrate that tau is found in the glomeruli of the renal cortex, and specifically in podocytes. This was further confirmed by immunohistochemistry. TauGFP/GFP mice present a podocyte cytoskeleton more dynamic as they contain higher levels of detyrosinated tubulin than wild-type mice. In addition, transmission electron microscopy studies demonstrated glomerular damage with a decrease in urinary creatinine. Our results prove that tau has an important role in kidney metabolism under normal physiological conditions.
Collapse
Affiliation(s)
- Laura Vallés-Saiz
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain
| | - Rocio Peinado-Cahuchola
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain
| | - Jesús Ávila
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
12
|
Mielcarska MB, Skowrońska K, Wyżewski Z, Toka FN. Disrupting Neurons and Glial Cells Oneness in the Brain-The Possible Causal Role of Herpes Simplex Virus Type 1 (HSV-1) in Alzheimer's Disease. Int J Mol Sci 2021; 23:ijms23010242. [PMID: 35008671 PMCID: PMC8745046 DOI: 10.3390/ijms23010242] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Current data strongly suggest herpes simplex virus type 1 (HSV-1) infection in the brain as a contributing factor to Alzheimer's disease (AD). The consequences of HSV-1 brain infection are multilateral, not only are neurons and glial cells damaged, but modifications also occur in their environment, preventing the transmission of signals and fulfillment of homeostatic and immune functions, which can greatly contribute to the development of disease. In this review, we discuss the pathological alterations in the central nervous system (CNS) cells that occur, following HSV-1 infection. We describe the changes in neurons, astrocytes, microglia, and oligodendrocytes related to the production of inflammatory factors, transition of glial cells into a reactive state, oxidative damage, Aβ secretion, tau hyperphosphorylation, apoptosis, and autophagy. Further, HSV-1 infection can affect processes observed during brain aging, and advanced age favors HSV-1 reactivation as well as the entry of the virus into the brain. The host activates pattern recognition receptors (PRRs) for an effective antiviral response during HSV-1 brain infection, which primarily engages type I interferons (IFNs). Future studies regarding the influence of innate immune deficits on AD development, as well as supporting the neuroprotective properties of glial cells, would reveal valuable information on how to harness cytotoxic inflammatory milieu to counter AD initiation and progression.
Collapse
Affiliation(s)
- Matylda Barbara Mielcarska
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-59-36063
| | - Katarzyna Skowrońska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Adolfa Pawińskiego 5, 02-106 Warsaw, Poland;
| | - Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University in Warsaw, Dewajtis 5, 01-815 Warsaw, Poland;
| | - Felix Ngosa Toka
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland;
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre 42123, Saint Kitts and Nevis
| |
Collapse
|
13
|
Wu M, Zhang M, Yin X, Chen K, Hu Z, Zhou Q, Cao X, Chen Z, Liu D. The role of pathological tau in synaptic dysfunction in Alzheimer's diseases. Transl Neurodegener 2021; 10:45. [PMID: 34753506 PMCID: PMC8579533 DOI: 10.1186/s40035-021-00270-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline, accompanied by amyloid-β (Aβ) overload and hyperphosphorylated tau accumulation in the brain. Synaptic dysfunction, an important pathological hallmark in AD, is recognized as the main cause of the cognitive impairments. Accumulating evidence suggests that synaptic dysfunction could be an early pathological event in AD. Pathological tau, which is detached from axonal microtubules and mislocalized into pre- and postsynaptic neuronal compartments, is suggested to induce synaptic dysfunction in several ways, including reducing mobility and release of presynaptic vesicles, decreasing glutamatergic receptors, impairing the maturation of dendritic spines at postsynaptic terminals, disrupting mitochondrial transport and function in synapses, and promoting the phagocytosis of synapses by microglia. Here, we review the current understanding of how pathological tau mediates synaptic dysfunction and contributes to cognitive decline in AD. We propose that elucidating the mechanism by which pathological tau impairs synaptic function is essential for exploring novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Moxin Wu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China.,Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China
| | - Manqing Zhang
- Medical College of Jiujiang University, Jiujiang, 332000, China
| | - Xiaoping Yin
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China.,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Kai Chen
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhijian Hu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Qin Zhou
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China
| | - Xianming Cao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China.,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China. .,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China.
| | - Dan Liu
- Department of Medical Genetics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
14
|
Vega AR, Chkheidze R, Jarmale V, Shang P, Foong C, Diamond MI, White CL, Rajaram S. Deep learning reveals disease-specific signatures of white matter pathology in tauopathies. Acta Neuropathol Commun 2021; 9:170. [PMID: 34674762 PMCID: PMC8529809 DOI: 10.1186/s40478-021-01271-x] [Citation(s) in RCA: 6] [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] [Received: 08/12/2021] [Accepted: 10/07/2021] [Indexed: 02/08/2023] Open
Abstract
Although pathology of tauopathies is characterized by abnormal tau protein aggregation in both gray and white matter regions of the brain, neuropathological investigations have generally focused on abnormalities in the cerebral cortex because the canonical aggregates that form the diagnostic criteria for these disorders predominate there. This corticocentric focus tends to deemphasize the relevance of the more complex white matter pathologies, which remain less well characterized and understood. We took a data-driven machine-learning approach to identify novel disease-specific morphologic signatures of white matter aggregates in three tauopathies: Alzheimer disease (AD), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD). We developed automated approaches using whole slide images of tau immunostained sections from 49 human autopsy brains (16 AD,13 CBD, 20 PSP) to identify cortex/white matter regions and individual tau aggregates, and compared tau-aggregate morphology across these diseases. Tau burden in the gray and white matter for individual subjects strongly correlated in a highly disease-specific fashion. We discovered previously unrecognized tau morphologies for AD, CBD and PSP that may be of importance in disease classification. Intriguingly, our models classified diseases equally well based on either white or gray matter tau staining. Our results suggest that tau pathology in white matter is informative, disease-specific, and linked to gray matter pathology. Machine learning has the potential to reveal latent information in histologic images that may represent previously unrecognized patterns of neuropathology, and additional studies of tau pathology in white matter could improve diagnostic accuracy.
Collapse
Affiliation(s)
- Anthony R Vega
- Lyda Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, USA
- Center for Alzheimer's and Neurodegenerative Diseases, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Rati Chkheidze
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
| | - Vipul Jarmale
- Lyda Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Ping Shang
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Chan Foong
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Marc I Diamond
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, USA
- Center for Alzheimer's and Neurodegenerative Diseases, The University of Texas Southwestern Medical Center, Dallas, USA
- Peter O'Donnell Jr. Brain Institute, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Charles L White
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, USA
- Center for Alzheimer's and Neurodegenerative Diseases, The University of Texas Southwestern Medical Center, Dallas, USA
- Peter O'Donnell Jr. Brain Institute, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Satwik Rajaram
- Lyda Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, USA.
- Center for Alzheimer's and Neurodegenerative Diseases, The University of Texas Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
15
|
Wang P, Ye Y. Astrocytes in Neurodegenerative Diseases: A Perspective from Tauopathy and α-Synucleinopathy. Life (Basel) 2021; 11:life11090938. [PMID: 34575087 PMCID: PMC8471224 DOI: 10.3390/life11090938] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases are aging-associated chronic pathological conditions affecting primarily neurons in humans. Inclusion bodies containing misfolded proteins have emerged as a common pathologic feature for these diseases. In many cases, misfolded proteins produced by a neuron can be transmitted to another neuron or a non-neuronal cell, leading to the propagation of disease-associated pathology. While undergoing intercellular transmission, misfolded proteins released from donor cells can often change the physiological state of recipient cells. Accumulating evidence suggests that astrocytes are highly sensitive to neuron-originated proteotoxic insults, which convert them into an active inflammatory state. Conversely, activated astrocytes can release a plethora of factors to impact neuronal functions. This review summarizes our current understanding of the complex molecular interplays between astrocyte and neuron, emphasizing on Tau and α-synuclein (α-syn), the disease-driving proteins for Alzheimer's and Parkinson's diseases, respectively.
Collapse
Affiliation(s)
| | - Yihong Ye
- Correspondence: ; Tel.: +1-301-594-0845; Fax: +1-301-496-0201
| |
Collapse
|
16
|
McCann H, Durand B, Shepherd CE. Aging-Related Tau Astrogliopathy in Aging and Neurodegeneration. Brain Sci 2021; 11:brainsci11070927. [PMID: 34356161 PMCID: PMC8306417 DOI: 10.3390/brainsci11070927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
Astrocytes are of vital importance to neuronal function and the health of the central nervous system (CNS), and astrocytic dysfunction as a primary or secondary event may predispose to neurodegeneration. Until recently, the main astrocytic tauopathies were the frontotemporal lobar degeneration with tau (FTLD-tau) group of disorders; however, aging-related tau astrogliopathy (ARTAG) has now been defined. This condition is a self-describing neuropathology mainly found in individuals over 60 years of age. Astrocytic tau accumulates with a thorny or granular/fuzzy morphology and is commonly found in normal aging as well as coexisting with diverse neurodegenerative disorders. However, there are still many unknown factors associated with ARTAG, including the cause/s, the progression, and the nature of any clinical associations. In addition to FTLD-tau, ARTAG has recently been associated with chronic traumatic encephalopathy (CTE), where it has been proposed as a potential precursor to these conditions, with the different ARTAG morphological subtypes perhaps having separate etiologies. This is an emerging area of exciting research that encompasses complex neurobiological and clinicopathological investigation.
Collapse
Affiliation(s)
- Heather McCann
- Neuroscience Research Australia, Barker Street, Sydney, NSW 2031, Australia; (H.M.); (B.D.)
| | - Briony Durand
- Neuroscience Research Australia, Barker Street, Sydney, NSW 2031, Australia; (H.M.); (B.D.)
| | - Claire E. Shepherd
- Neuroscience Research Australia, Barker Street, Sydney, NSW 2031, Australia; (H.M.); (B.D.)
- Department of Pathology, The University of New South Wales, Kensington, Sydney, NSW 2031, Australia
- Correspondence:
| |
Collapse
|
17
|
Kanaan NM, Grabinski T. Neuronal and Glial Distribution of Tau Protein in the Adult Rat and Monkey. Front Mol Neurosci 2021; 14:607303. [PMID: 33986642 PMCID: PMC8112591 DOI: 10.3389/fnmol.2021.607303] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/23/2021] [Indexed: 12/27/2022] Open
Abstract
Tau is a microtubule-associated protein for which the physiological functions remain a topic of vigorous investigation. Additionally, tau is a central player in the pathogenesis of several diseases such as Alzheimer's disease and several frontotemporal dementias. A critical variable to understanding tau in physiological and disease contexts is its normal localization within cells of the adult CNS. Tau is often described as an axon-specific (or enriched) and neuron-specific protein with little to no expression in glial cells, all of which are untrue. Understanding normal tau distribution also impacts interpretation of experimental results and hypotheses regarding its role in disease. Thus, we set out to help clarify the normal localization of tau in the adult CNS of middle-aged rats and rhesus macaque using the hippocampus as a representative brain structure. The physiological concentration of tau in the rat hippocampus was 6.6 μM and in white matter was 3.6 μM as determined by quantitative sandwich ELISAs. We evaluated the cellular localization of tau using multiple tau-specific antibodies with epitopes to different regions, including Tau1, Tau5, Tau7, R1, and two novel primate-specific antibodies NT9 and NT15. In the rat and monkey, tau was localized within the somatodendritic and axonal compartments, as well as a subset of neuronal nuclei. Semi-quantitative fluorescence intensity measurements revealed that depending on the specific reagent used the somatodendritic tau is relatively equal to, higher than, or lower than axonal tau, highlighting differential labeling of tau with various antibodies despite its distribution throughout the neuron. Tau was strongly expressed in mature oligodendrocytes and displayed little to no expression in oligodendrocyte precursor cells, astrocytes or microglia. Collectively, the data indicate tau is ∼3 - 7 μM under physiological conditions, is not specifically enriched in axons, and is normally found in both neurons and mature oligodendrocytes in the adult CNS. The full landscape of tau distribution is not revealed by all antibodies suggesting availability of the epitopes is different within specific neuronal compartments. These findings set the stage for better understanding normal tau distributions and interpreting data regarding the presence of tau in different compartments or cell types within disease conditions.
Collapse
Affiliation(s)
- Nicholas M. Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Neuroscience Program, Michigan State University, East Lansing, MI, United States
- Mercy Health Hauenstein Neuroscience Center, Grand Rapids, MI, United States
| | - Tessa Grabinski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| |
Collapse
|
18
|
Criado-Marrero M, Gebru NT, Blazier DM, Gould LA, Baker JD, Beaulieu-Abdelahad D, Blair LJ. Hsp90 co-chaperones, FKBP52 and Aha1, promote tau pathogenesis in aged wild-type mice. Acta Neuropathol Commun 2021; 9:65. [PMID: 33832539 PMCID: PMC8033733 DOI: 10.1186/s40478-021-01159-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
The microtubule associated protein tau is an intrinsically disordered phosphoprotein that accumulates under pathological conditions leading to formation of neurofibrillary tangles, a hallmark of Alzheimer's disease (AD). The mechanisms that initiate the accumulation of phospho-tau aggregates and filamentous deposits are largely unknown. In the past, our work and others' have shown that molecular chaperones play a crucial role in maintaining protein homeostasis and that imbalance in their levels or activity can drive tau pathogenesis. We have found two co-chaperones of the 90 kDa heat shock protein (Hsp90), FK506-binding protein 52 (FKBP52) and the activator of Hsp90 ATPase homolog 1 (Aha1), promote tau aggregation in vitro and in the brains of tau transgenic mice. Based on this, we hypothesized that increased levels of these chaperones could promote tau misfolding and accumulation in the brains of aged wild-type mice. We tested this hypothesis by overexpressing Aha1, FKBP52, or mCherry (control) proteins in the hippocampus of 9-month-old wild-type mice. After 7 months of expression, mice were evaluated for cognitive and pathological changes. Our results show that FKBP52 overexpression impaired spatial reversal learning, while Aha1 overexpression impaired associative learning in aged wild-type mice. FKBP52 and Aha1 overexpression promoted phosphorylation of distinct AD-relevant tau species. Furthermore, FKBP52 activated gliosis and promoted neuronal loss leading to a reduction in hippocampal volume. Glial activation and phospho-tau accumulation were also detected in areas adjacent to the hippocampus, including the entorhinal cortex, suggesting that after initiation these pathologies can propagate through other brain regions. Overall, our findings suggest a role for chaperone imbalance in the initiation of tau accumulation in the aging brain.
Collapse
Affiliation(s)
- Marangelie Criado-Marrero
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA
| | - Niat T Gebru
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA
| | - Danielle M Blazier
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA
| | - Lauren A Gould
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA
| | - Jeremy D Baker
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA
| | - David Beaulieu-Abdelahad
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA
| | - Laura J Blair
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA.
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA.
- Research Service, James A Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
19
|
Fleeman RM, Proctor EA. Astrocytic Propagation of Tau in the Context of Alzheimer's Disease. Front Cell Neurosci 2021; 15:645233. [PMID: 33815065 PMCID: PMC8010320 DOI: 10.3389/fncel.2021.645233] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/19/2021] [Indexed: 01/14/2023] Open
Abstract
More than 6 million Americans are currently living with Alzheimer's disease (AD), and the incidence is growing rapidly with our aging population. Numerous therapeutics have failed to make it to the clinic, potentially due to a focus on presumptive pathogenic proteins instead of cell-type-specific signaling mechanisms. The tau propagation hypothesis that inter-neuronal tau transfer drives AD pathology has recently garnered attention, as accumulation of pathological tau in the brain has high clinical significance in correlating with progression of cognitive AD symptoms. However, studies on tau pathology in AD are classically neuron-centric and have greatly overlooked cell-type specific effects of tau internalization, degradation, and propagation. While the contribution of microglia to tau processing and propagation is beginning to be recognized and understood, astrocytes, glial cells in the brain important for maintaining neuronal metabolic, synaptic, trophic, and immune function which can produce, internalize, degrade, and propagate tau are understudied in their ability to affect AD progression through tau pathology. Here, we showcase evidence for whether tau uptake by astrocytes may be beneficial or detrimental to neuronal health and how astrocytes and their immunometabolic functions may be key targets for future successful AD therapies.
Collapse
Affiliation(s)
- Rebecca M Fleeman
- Department of Neurosurgery, Department of Pharmacology, College of Medicine, Pennsylvania State University (PSU), Hershey, PA, United States.,Center for Neural Engineering, Pennsylvania State University (PSU), University Park, PA, United States
| | - Elizabeth A Proctor
- Department of Neurosurgery, Department of Pharmacology, College of Medicine, Pennsylvania State University (PSU), Hershey, PA, United States.,Department of Biomedical Engineering, Department of Engineering Science and Mechanics, Center for Neural Engineering, Pennsylvania State University (PSU), University Park, PA, United States
| |
Collapse
|
20
|
Song L, Wells EA, Robinson AS. Critical Molecular and Cellular Contributors to Tau Pathology. Biomedicines 2021; 9:190. [PMID: 33672982 PMCID: PMC7918468 DOI: 10.3390/biomedicines9020190] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/07/2021] [Accepted: 02/11/2021] [Indexed: 12/11/2022] Open
Abstract
Tauopathies represent a group of neurodegenerative diseases including Alzheimer's disease (AD) that are characterized by the deposition of filamentous tau aggregates in the brain. The pathogenesis of tauopathies starts from the formation of toxic 'tau seeds' from hyperphosphorylated tau monomers. The presence of specific phosphorylation sites and heat shock protein 90 facilitates soluble tau protein aggregation. Transcellular propagation of pathogenic tau into synaptically connected neuronal cells or adjacent glial cells via receptor-mediated endocytosis facilitate disease spread through the brain. While neuroprotective effects of glial cells-including phagocytotic microglial and astroglial phenotypes-have been observed at the early stage of neurodegeneration, dysfunctional neuronal-glial cellular communication results in a series of further pathological consequences as the disease progresses, including abnormal axonal transport, synaptic degeneration, and neuronal loss, accompanied by a pro-inflammatory microenvironment. Additionally, the discovery of microtubule-associated protein tau (MAPT) gene mutations and the strongest genetic risk factor of tauopathies-an increase in the presence of the ε2 allele of apolipoprotein E (ApoE)-provide important clues to understanding tau pathology progression. In this review, we describe the crucial signaling pathways and diverse cellular contributors to the progression of tauopathies. A systematic understanding of disease pathogenesis provides novel insights into therapeutic targets within altered signaling pathways and is of great significance for discovering effective treatments for tauopathies.
Collapse
Affiliation(s)
| | | | - Anne Skaja Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (L.S.); (E.A.W.)
| |
Collapse
|
21
|
Zareba-Paslawska J, Patra K, Kluzer L, Revesz T, Svenningsson P. Tau Isoform-Driven CBD Pathology Transmission in Oligodendrocytes in Humanized Tau Mice. Front Neurol 2021; 11:589471. [PMID: 33519674 PMCID: PMC7845573 DOI: 10.3389/fneur.2020.589471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
The aggregation of abnormally phosphorylated tau protein in neurons and glia is a neuropathological hallmark of several neurodegenerative disorders, collectively known as tauopathies. They are further subclassified based on the preferential pathological aggregation of three carboxyl-terminal repeat domains (3R) and/or 4R tau. Corticobasal degeneration (CBD) is a rare neurodegenerative disorder classified as a 4R tauopathy. In the present study, we extend analysis of CBD-tau cell-type specific pathology transmission with 3R and 4R tau isoform distinguishable changes. We use a humanized tau (hTau) mouse line, which overexpress all six human tau isoforms in a murine tau knockout background and perform intrastriatal inoculation of control and CBD-tau enriched human brain homogenate. We show that CBD-tau causes hyperphosphorylation of tau at Ser202 predominantly in oligodendrocytes. Next, we demonstrate the spread of tau pathology from striatum to the overlaying corpus callosum and further to the contralateral side. Finally, we demonstrate that the almost exclusive oligodendrocyte-based transmission of hyperphosphorylated tau is reflected in the endogenous 4R tau isoform expression and corresponds to subclassification of CBD as a 4R tauopathy. Additionally, we identify functional changes in oligodendrocytes reflected by myelin basic protein abnormalities upon CBD-tau inoculation. These changes are not observed in murine tau knockout mice lacking both human and murine tau. Our study presents not only in vivo tau isoform–driven region- and cell-specific tau pathology, but also underlines that tau pathology seeding and transmission might be oligodendrocyte-based. These results, which need to be extended to more cases, give new insights into why tauopathies might vary greatly in both histopathological and neuroanatomical patterns.
Collapse
Affiliation(s)
- Justyna Zareba-Paslawska
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Kalicharan Patra
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Luca Kluzer
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Tamas Revesz
- Queen Square Brain Bank, Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Per Svenningsson
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
22
|
Wang P, Ye Y. Filamentous recombinant human Tau activates primary astrocytes via an integrin receptor complex. Nat Commun 2021; 12:95. [PMID: 33398028 PMCID: PMC7782792 DOI: 10.1038/s41467-020-20322-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022] Open
Abstract
Microtubule-associated protein Tau can form protein aggregates transmissible within the brain, correlating with the progression of tauopathies in humans. The transmission of aggregates requires neuron-released Tau to interact with surface receptors on target cells. However, the underlying molecular mechanisms in astrocytes and downstream effects are unclear. Here, using a spatially resolved proteomic mapping strategy, we show that integrin αV/β1 receptor binds recombinant human Tau, mediating the entry of Tau fibrils in astrocytes. The binding of distinct Tau species to the astrocytic αV/β1 receptor differentially activate integrin signaling. Furthermore, Tau-mediated activation of integrin signaling results in NFκB activation, causing upregulation of pro-inflammatory cytokines and chemokines, induction of a sub-group of neurotoxic astrocytic markers, and release of neurotoxic factors. Our findings suggest that filamentous recombinant human Tau-mediated activation of integrin signaling induces astrocyte conversion towards a neurotoxic state, providing a mechanistic insight into tauopathies.
Collapse
Affiliation(s)
- Peng Wang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
23
|
Bourojeni FB, Zeilhofer HU, Kania A. Netrin-1 receptor DCC is required for the contralateral topography of lamina I anterolateral system neurons. Pain 2021; 162:161-175. [PMID: 32701653 PMCID: PMC7737868 DOI: 10.1097/j.pain.0000000000002012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 12/30/2022]
Abstract
Anterolateral system (AS) neurons relay nociceptive information from the spinal cord to the brain, protecting the body from harm by evoking a variety of behaviours and autonomic responses. The developmental programs that guide the connectivity of AS neurons remain poorly understood. Spinofugal axons cross the spinal midline in response to Netrin-1 signalling through its receptor deleted in colorectal carcinoma (DCC); however, the relevance of this canonical pathway to AS neuron development has only been demonstrated recently. Here, we disrupted Netrin-1:DCC signalling developmentally in AS neurons and assessed the consequences on the path finding of the different classes of spinofugal neurons. Many lamina I AS neurons normally innervate the lateral parabrachial nucleus and periaqueductal gray on the contralateral side. The loss of DCC in the developing spinal cord resulted in increased frequency of ipsilateral projection of spinoparabrachial and spinoperiaqueductal gray neurons. Given that contralateral spinofugal projections are largely associated with somatotopic representation of the body, changes in the laterality of AS spinofugal projections may contribute to reduced precision in pain localization observed in mice and humans carrying Dcc mutations.
Collapse
Affiliation(s)
- Farin B. Bourojeni
- Research Unit in Neural Circuit Development, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Switzerland
| | - Artur Kania
- Research Unit in Neural Circuit Development, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Division of Experimental Medicine, Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| |
Collapse
|
24
|
Does a hypoxic injury from a non-fatal overdose lead to an Alzheimer Disease? Neurochem Int 2020; 143:104936. [PMID: 33309980 DOI: 10.1016/j.neuint.2020.104936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/21/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022]
Abstract
Long term consequence of non-fatal overdose in people who use opioids are not well understood. The intermittent exposure to non-fatal overdose leads to a tauopathy that is often accompanied by abrogated neuroprotective response, abnormal amyloid processing and other pathologies. The scope and limitations of available literature are discussed including neuropathologies associated with opioid and overdose exposures, contributing comorbidities and proteinopathies. Contrasting postmortem data of overdose victims with animal models of opioid neuropathologies and hypoxic injury paints a picture distinct from other proteinopathies as well as effects of moderate opioid exposure. Furthermore the reported biochemical changes and potential targets for therapeutic intervention were mapped pointing to underlying imbalance between tau kinases and phosphatases that is characteristic of Alzheimer Disease.
Collapse
|
25
|
Merezhko M, Uronen RL, Huttunen HJ. The Cell Biology of Tau Secretion. Front Mol Neurosci 2020; 13:569818. [PMID: 33071756 PMCID: PMC7539664 DOI: 10.3389/fnmol.2020.569818] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022] Open
Abstract
The progressive accumulation and spread of misfolded tau protein in the nervous system is the hallmark of tauopathies, progressive neurodegenerative diseases with only symptomatic treatments available. A growing body of evidence suggests that spreading of tau pathology can occur via cell-to-cell transfer involving secretion and internalization of pathological forms of tau protein followed by templated misfolding of normal tau in recipient cells. Several studies have addressed the cell biological mechanisms of tau secretion. It now appears that instead of a single mechanism, cells can secrete tau via three coexisting pathways: (1) translocation through the plasma membrane; (2) membranous organelles-based secretion; and (3) ectosomal shedding. The relative importance of these pathways in the secretion of normal and pathological tau is still elusive, though. Moreover, glial cells contribute to tau propagation, and the involvement of different cell types, as well as different secretion pathways, complicates the understanding of prion-like propagation of tauopathy. One of the important regulators of tau secretion in neuronal activity, but its mechanistic connection to tau secretion remains unclear and may involve all three secretion pathways of tau. This review article summarizes recent advancements in the field of tau secretion with an emphasis on cell biological aspects of the secretion process and discusses the role of neuronal activity and glial cells in the spread of pathological forms of tau.
Collapse
Affiliation(s)
- Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
Zubčić K, Hof PR, Šimić G, Jazvinšćak Jembrek M. The Role of Copper in Tau-Related Pathology in Alzheimer's Disease. Front Mol Neurosci 2020; 13:572308. [PMID: 33071757 PMCID: PMC7533614 DOI: 10.3389/fnmol.2020.572308] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
All tauopathies, including Alzheimer's disease (AD), are characterized by the intracellular accumulation of abnormal forms of tau protein in neurons and glial cells, which negatively affect microtubule stability. Under physiological conditions, tubulin-associated unit (Tau) protein is intrinsically disordered, almost without secondary structure, and is not prone to aggregation. In AD, it assembles, and forms paired helical filaments (PHFs) that further build-up neurofibrillary tangles (NFTs). Aggregates are composed of hyperphosphorylated tau protein that is more prone to aggregation. The pathology of AD is also linked to disturbed copper homeostasis, which promotes oxidative stress (OS). Copper imbalance is widely observed in AD patients. Deregulated copper ions may initiate and exacerbate tau hyperphosphorylation and formation of β-sheet-rich tau fibrils that ultimately contribute to synaptic failure, neuronal death, and cognitive decline observed in AD patients. The present review summarizes factors affecting the process of tau aggregation, conformational changes of small peptide sequences in the microtubule-binding domain required for these motifs to act as seeding sites in aggregation, and the role of copper in OS induction, tau hyperphosphorylation and tau assembly. A better understanding of the various factors that affect tau aggregation under OS conditions may reveal new targets and novel pharmacological approaches for the therapy of AD.
Collapse
Affiliation(s)
- Klara Zubčić
- Laboratory for Developmental Neuropathology, Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Laboratory for Developmental Neuropathology, Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Maja Jazvinšćak Jembrek
- Laboratory for Protein Dynamics, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia.,Department of Psychology, Catholic University of Croatia, Zagreb, Croatia
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Progressive supranuclear palsy (PSP) is a progressive adult-onset neurodegenerative disease. Abnormally, phosphorylated forms of the microtubule-associated protein tau containing four repeat domains (4R-tau) aggregate in neurons. Additionally, increasing evidence suggests that secretion and uptake of fragments of abnormal 4R-tau may play a role in disease progression. This extracellular tau is a natural target for immunotherapy. RECENT FINDINGS Three monoclonal antibodies targeting extracellular tau are in clinical stages of development. ABBV-8E12 and BIIB092 were safe in Phase 1, but both Phase two studies recently failed futility analyses. UCB0107 recently reported (in abstract form) Phase 1 safety results, and a Phase 2 study is under consideration. Stem cell therapy and the infusion of plasma are also being explored clinically. SUMMARY The likely role of extracellular tau in the progression of PSP makes tau a natural target for targeted immunotherapy. Clinical trials are still in early stages, and although tau immunotherapy has largely been shown to be safe, efficacy has yet to be demonstrated.
Collapse
Affiliation(s)
- Pavan A Vaswani
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
28
|
Means JC, Lopez AA, Koulen P. Resveratrol Protects Optic Nerve Head Astrocytes from Oxidative Stress-Induced Cell Death by Preventing Caspase-3 Activation, Tau Dephosphorylation at Ser 422 and Formation of Misfolded Protein Aggregates. Cell Mol Neurobiol 2020; 40:911-926. [PMID: 31919747 PMCID: PMC7299779 DOI: 10.1007/s10571-019-00781-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022]
Abstract
Optic nerve head astrocytes (ONHAs) are the major cell type within the optic nerve head, providing both structural and nutrient support to the optic nerve. Astrocytes are necessary for the survival of neurons with controlled activation of astrocytes being beneficial to neurons. However, overactive astrocytes can be harmful and the loss of normal astrocyte function can be a primary contributor to neurodegeneration. The neuroprotective properties of reactive astrocytes can be lost or they might gain neurotoxic properties in neurodegenerative diseases. The activated astrocytes are crucial in the development of glaucoma, where they serve as a source for cytotoxic substances that participate in ganglion apoptosis. There is increasing evidence indicating that neuroinflammation is an important process in glaucoma. Under pathological conditions, astrocytes can induce an inflammatory response. Extensive evidence shows that inflammatory responses mediated by astrocytes can also influence pathology development, synapse health, and neurodegeneration. The elimination of activated astrocytes by apoptosis is also expected in unfavorable conditions. In neurodegenerative diseases, a common feature is the presence of aggregates found in astrocytes, which can disrupt astrocyte function in such a way as to be detrimental to the viability of neurons. The biological processes involved in vision loss in glaucoma are not well understood. Despite the rapid advances in our understanding of optic nerve head (ONH) structure and function, numerous potential contributions of the ONHAs to optic nerve damage remain unanswered. The present study investigated the role of ONHAs during oxidative stress in order to determine novel cell biological processes underlying glaucoma pathogenesis. ONHAs were exposed to chemically induced oxidative stress using tert-butyl hydroperoxide (tBHP) in order to model extracellular oxidative stress as it occurs in the glaucomatous retina and ONH. In order to determine the impact of an intervention approach employing potential glioprotective treatments for central nervous system tissue we pretreated cells with the polyphenolic phytostilbene and antioxidant trans-resveratrol (3,5,4'-trihydroxy-trans-stilbene). ONHAs exposed to tBHP-mediated oxidative stress displayed decreased viability and underwent apoptosis. In addition, increased levels of activated caspases, dephosphorylation of Tau protein at Ser422, an important site adjacent to the caspase cleavage site controlling Tau cleavage, caspase-mediated Tau cleavage, and cytoskeletal changes, specifically formation of neurofibrillary tangles (NFTs) were detected in ONHAs undergoing oxidative stress. When cells were pretreated with resveratrol cell viability increased along with a significant decrease in activated caspases, cleaved Tau, and NFT formation. Taken together, ONHAs appear to act similar to neurons when undergoing oxidative stress, where proteolytic cleavage of Tau by caspases leads to NFT formation. In addition, resveratrol appears to have promise as a potential protective treatment preventing ONHA dysfunction and degeneration. There is currently no cure for glaucoma or a neuro- and glioprotective treatment that directly targets the pathogenic mechanisms in the glaucomatous retina and optic nerve. The present study identified a potential mechanism underlying degeneration of astrocytes that is susceptible to pharmaco-therapeutic intervention in the eye and potentially elsewhere in the central nervous system. Identification of such mechanisms involved in glaucoma and other disorders of the eye and brain is critical to determine novel targets for effective therapies.
Collapse
Affiliation(s)
- John C Means
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri -Kansas City, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Adam A Lopez
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri -Kansas City, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Peter Koulen
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri -Kansas City, 2411 Holmes St, Kansas City, MO, 64108, USA.
- Department of Biomedical Sciences, School of Medicine, University of Missouri -Kansas City, 2411 Holmes St, Kansas City, MO, 64108, USA.
| |
Collapse
|
29
|
Wong CO. Endosomal-Lysosomal Processing of Neurodegeneration-Associated Proteins in Astrocytes. Int J Mol Sci 2020; 21:ijms21145149. [PMID: 32708198 PMCID: PMC7404029 DOI: 10.3390/ijms21145149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/15/2022] Open
Abstract
Most common neurodegenerative diseases (NDs) are characterized by deposition of protein aggregates that are resulted from misfolding, dysregulated trafficking, and compromised proteolytic degradation. These proteins exert cellular toxicity to a broad range of brain cells and are found in both neurons and glia. Extracellular monomeric and oligomeric ND-associated proteins are taken up by astrocytes, the most abundant glial cell in the brain. Internalization, intracellular trafficking, processing, and disposal of these proteins are executed by the endosomal-lysosomal system of astrocytes. Endosomal-lysosomal organelles thus mediate the cellular impact and metabolic fate of these toxic protein species. Given the indispensable role of astrocytes in brain metabolic homeostasis, the endosomal-lysosomal processing of these proteins plays a fundamental role in altering the trajectory of neurodegeneration. This review aims at summarizing the mounting evidence that has established the essential role of astrocytic endosomal-lysosomal organelles in the processing of amyloid precursor proteins, Apolipoprotein E (ApoE), tau, alpha synuclein, and huntingtin, which are associated with NDs such as Alzheimer’s, Parkinson’s, and Huntington diseases.
Collapse
Affiliation(s)
- Ching-On Wong
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
30
|
LeBlang CJ, Medalla M, Nicoletti NW, Hays EC, Zhao J, Shattuck J, Cruz AL, Wolozin B, Luebke JI. Reduction of the RNA Binding Protein TIA1 Exacerbates Neuroinflammation in Tauopathy. Front Neurosci 2020; 14:285. [PMID: 32327969 PMCID: PMC7161592 DOI: 10.3389/fnins.2020.00285] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammatory processes play an integral role in the exacerbation and progression of pathology in tauopathies, a class of neurodegenerative disease characterized by aggregation of hyperphosphorylated tau protein. The RNA binding protein (RBP) T-cell Intracellular Antigen 1 (TIA1) is an important regulator of the innate immune response in the periphery, dampening cytotoxic inflammation and apoptosis during cellular stress, however, its role in neuroinflammation is unknown. We have recently shown that TIA1 regulates tau pathophysiology and toxicity in part through the binding of phospho-tau oligomers into pathological stress granules, and that haploinsufficiency of TIA1 in the P301S mouse model of tauopathy results in reduced accumulation of toxic tau oligomers, pathologic stress granules, and the development of downstream pathological features of tauopathy. The putative role of TIA1 as a regulator of the peripheral immune response led us to investigate the effects of TIA1 on neuroinflammation in the context of tauopathy, a chronic stressor in the neural environment. Here, we evaluated indicators of neuroinflammation including; reactive microgliosis and phagocytosis, pro-inflammatory cytokine release, and oxidative stress in hippocampal neurons and glia of wildtype and P301S transgenic mice expressing TIA1+/+, TIA1+/-, and TIA1-/- in both early (5 month) and advanced (9 month) disease states through biochemical, ultrastructural, and histological analyses. Our data show that both TIA1 haploinsufficiency and TIA1 knockout exacerbate neuroinflammatory processes in advanced stages of tauopathy, suggesting that TIA1 dampens the immune response in the central nervous system during chronic stress.
Collapse
Affiliation(s)
- Chelsey Jenna LeBlang
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Maria Medalla
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Nicholas William Nicoletti
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Emma Catherine Hays
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - James Zhao
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Jenifer Shattuck
- Laboratory of Neurodegeneration, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Anna Lourdes Cruz
- Laboratory of Neurodegeneration, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Benjamin Wolozin
- Laboratory of Neurodegeneration, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- Department of Neuroscience, Boston University, Boston, MA, United States
| | - Jennifer Irene Luebke
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
31
|
Abstract
In some neurodegenerative disorders, tau protein accumulates in astrocytes and/or oligodendrocytes, even though these glial cells produce much less of the protein than do neurons. Testing the hypothesis that the aggregated tau in glia derives from neurons, Narasimhan et al. (https://doi.org/10.1084/jem.20190783) make the unexpected discovery that neuronal tau expression is not required for the formation of glial tau inclusions. The circumstances governing the variable cell-specificity of tauopathy thus remain to be fully defined.
Collapse
Affiliation(s)
- Lary C. Walker
- Department of Neurology and Yerkes National Primate Research Center, Emory University, Atlanta, GA
| |
Collapse
|
32
|
Resende EDPF, Nolan AL, Petersen C, Ehrenberg AJ, Spina S, Allen IE, Rosen HJ, Kramer J, Miller BL, Seeley WW, Gorno-Tempini ML, Miller Z, Grinberg LT. Language and spatial dysfunction in Alzheimer disease with white matter thorn-shaped astrocytes. Neurology 2020; 94:e1353-e1364. [PMID: 32001514 DOI: 10.1212/wnl.0000000000008937] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Alzheimer disease (AD) shows a broad array of clinical presentations, but the mechanisms underlying these phenotypic variants remain elusive. Aging-related astrogliopathy (ARTAG) is a relatively recent term encompassing a broad array of tau deposition in astroglia outside the range of traditional tauopathies. White matter thorn-shaped astrocyte (WM-TSA) clusters, a specific ARTAG subtype, has been associated with atypical language presentation of AD in a small study lacking replication. To interrogate the impact of WM-TSA in modifying clinical phenotype in AD, we investigated a clinicopathologic sample of 83 persons with pure cortical AD pathology and heterogeneous clinical presentations. METHODS We mapped WM-TSA presence and density throughout cortical areas and interrogated whether WM-TSA correlated with atypical AD presentation or worse performance in neuropsychological testing. RESULTS WM-TSA was present in nearly half of the cases and equally distributed in typical and atypical AD presentations. Worsening language and visuospatial functions were correlated with higher WM-TSA density in language-related and visuospatial-related regions, respectively. These findings were unrelated to regional neurofibrillary tangle burden. Next, unsupervised clustering divided the participants into 2 groups: a high-WM-TSA (n = 9) and low-WM-TSA (n = 74) pathology signature. The high-WM-TSA group scored significantly worse in language but not in other cognitive domains. CONCLUSIONS The negative impact of WM-TSA pathology to language and possibly visuospatial networks suggests that WM-TSA is not as benign as other ARTAG types and may be explored as a framework to understand the mechanisms and impact of astrocytic tau deposition in AD in humans.
Collapse
Affiliation(s)
- Elisa de Paula França Resende
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Amber L Nolan
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Cathrine Petersen
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Alexander J Ehrenberg
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Salvatore Spina
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Isabel E Allen
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Howard J Rosen
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Joel Kramer
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Bruce L Miller
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - William W Seeley
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Maria Luiza Gorno-Tempini
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Zachary Miller
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Lea T Grinberg
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil.
| |
Collapse
|
33
|
White Matter and Neuroprotection in Alzheimer's Dementia. Molecules 2020; 25:molecules25030503. [PMID: 31979414 PMCID: PMC7038211 DOI: 10.3390/molecules25030503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Myelin is the main component of the white matter of the central nervous system (CNS), allowing the proper electrical function of the neurons by ensheathing and insulating the axons. The extensive use of magnetic resonance imaging has highlighted the white matter alterations in Alzheimer’s dementia (AD) and other neurodegenerative diseases, alterations which are early, extended, and regionally selective. Given that the white matter turnover is considerable in the adulthood, and that myelin repair is currently recognized as being the only true reparative capability of the mature CNS, oligodendrocyte precursor cells (OPCs), the cells that differentiate in oligodendrocyte, responsible for myelin formation and repair, are regarded as a potential target for neuroprotection. In this review, several aspects of the OPC biology are reviewed. The histology and functional role of OPCs in the neurovascular-neuroglial unit as described in preclinical and clinical studies on AD is discussed, such as the OPC vulnerability to hypoxia-ischemia, neuroinflammation, and amyloid deposition. Finally, the position of OPCs in drug discovery strategies for dementia is discussed.
Collapse
|
34
|
Shin J, Kim HJ, Jeon B. Immunotherapy Targeting Neurodegenerative Proteinopathies: α-Synucleinopathies and Tauopathies. J Mov Disord 2019; 13:11-19. [PMID: 31847513 PMCID: PMC6987523 DOI: 10.14802/jmd.19057] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022] Open
Abstract
α-Synuclein and tau deposition in the central nervous system is responsible for various parkinsonian syndromes, including Parkinson’s disease, multiple system atrophy, dementia with Lewy bodies, progressive supranuclear palsy and corticobasal degeneration. Emerging evidence has suggested that pathologic α-synuclein and tau are transmitted from cell to cell and further accelerate the aggregation of pathologic proteins in neighboring cells. Furthermore, extracellular pathologic proteins have also been reported to provoke inflammatory responses that lead to neurodegeneration. Therefore, immunotherapies targeting extracellular α-synuclein and tau have been proposed as potential disease-modifying strategies. In this review, we summarize completed phase I trials and ongoing phase II trials of immunotherapies against α-synuclein and tau and further discuss concerns and hurdles to overcome in the future.
Collapse
Affiliation(s)
- Junghwan Shin
- Department of Neurology and Movement Disorder Center, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Han-Joon Kim
- Department of Neurology and Movement Disorder Center, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Beomseok Jeon
- Department of Neurology and Movement Disorder Center, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Scarpelli EM, Trinh VY, Tashnim Z, Krans JL, Keller LC, Colodner KJ. Developmental expression of human tau in Drosophila melanogaster glial cells induces motor deficits and disrupts maintenance of PNS axonal integrity, without affecting synapse formation. PLoS One 2019; 14:e0226380. [PMID: 31821364 PMCID: PMC6903755 DOI: 10.1371/journal.pone.0226380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/25/2019] [Indexed: 11/26/2022] Open
Abstract
Tauopathies are a class of neurodegenerative diseases characterized by the abnormal phosphorylation and accumulation of the microtubule-associated protein, tau, in both neuronal and glial cells. Though tau pathology in glial cells is a prominent feature of many of these disorders, the pathological contribution of these lesions to tauopathy pathogenesis remains largely unknown. Moreover, while tau pathology is predominantly found in the central nervous system, a role for tau in the cells of the peripheral nervous system has been described, though not well characterized. To investigate the effects of glial tau expression on the development and maintenance of the peripheral nervous system, we utilized a Drosophila melanogaster model of tauopathy that expresses human wild-type tau in glial cells during development. We found that glial tau expression during development results in larval locomotor deficits and organismal lethality at the pupal stage, without affecting larval neuromuscular junction synapse development or post-synaptic amplitude. There was, however, a significant decrease in the decay time of synaptic potentials upon repeated stimulation of the motoneuron. Behavioral abnormalities were accompanied by glial cell death, disrupted maintenance of glial-axonal integrity, and the abnormal accumulation of the presynaptic protein, Bruchpilot, in peripheral nerve axons. Together, these data demonstrate that human tau expression in Drosophila glial cells does not affect neuromuscular junction synapse formation during development, but is deleterious to the maintenance of glial-axonal interactions in the peripheral nervous system.
Collapse
Affiliation(s)
- Enrico M. Scarpelli
- Frank H. Netter, M.D. School of Medicine, Quinnipiac University, North Haven, CT, United States of America
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, United States of America
| | - Van Y. Trinh
- Program in Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, United States of America
| | - Zarrin Tashnim
- Program in Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, United States of America
| | - Jacob L. Krans
- Department of Neuroscience, Western New England University, Springfield, MA, United States of America
| | - Lani C. Keller
- Frank H. Netter, M.D. School of Medicine, Quinnipiac University, North Haven, CT, United States of America
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, United States of America
| | - Kenneth J. Colodner
- Program in Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, United States of America
| |
Collapse
|
36
|
Proteasome Activation to Combat Proteotoxicity. Molecules 2019; 24:molecules24152841. [PMID: 31387243 PMCID: PMC6696185 DOI: 10.3390/molecules24152841] [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: 06/24/2019] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Loss of proteome fidelity leads to the accumulation of non-native protein aggregates and oxidatively damaged species: hallmarks of an aged cell. These misfolded and aggregated species are often found, and suggested to be the culpable party, in numerous neurodegenerative diseases including Huntington's, Parkinson's, Amyotrophic Lateral Sclerosis (ALS), and Alzheimer's Diseases (AD). Many strategies for therapeutic intervention in proteotoxic pathologies have been put forth; one of the most promising is bolstering the efficacy of the proteasome to restore normal proteostasis. This strategy is ideal as monomeric precursors and oxidatively damaged proteins, so called "intrinsically disordered proteins" (IDPs), are targeted by the proteasome. This review will provide an overview of disorders in proteins, both intrinsic and acquired, with a focus on susceptibility to proteasomal degradation. We will then examine the proteasome with emphasis on newly published structural data and summarize current known small molecule proteasome activators.
Collapse
|
37
|
Signaevsky M, Prastawa M, Farrell K, Tabish N, Baldwin E, Han N, Iida MA, Koll J, Bryce C, Purohit D, Haroutunian V, McKee AC, Stein TD, White CL, Walker J, Richardson TE, Hanson R, Donovan MJ, Cordon-Cardo C, Zeineh J, Fernandez G, Crary JF. Artificial intelligence in neuropathology: deep learning-based assessment of tauopathy. J Transl Med 2019; 99:1019-1029. [PMID: 30770886 PMCID: PMC7684013 DOI: 10.1038/s41374-019-0202-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 11/09/2022] Open
Abstract
Accumulation of abnormal tau in neurofibrillary tangles (NFT) occurs in Alzheimer disease (AD) and a spectrum of tauopathies. These tauopathies have diverse and overlapping morphological phenotypes that obscure classification and quantitative assessments. Recently, powerful machine learning-based approaches have emerged, allowing the recognition and quantification of pathological changes from digital images. Here, we applied deep learning to the neuropathological assessment of NFT in postmortem human brain tissue to develop a classifier capable of recognizing and quantifying tau burden. The histopathological material was derived from 22 autopsy brains from patients with tauopathies. We used a custom web-based informatics platform integrated with an in-house information management system to manage whole slide images (WSI) and human expert annotations as ground truth. We utilized fully annotated regions to train a deep learning fully convolutional neural network (FCN) implemented in PyTorch against the human expert annotations. We found that the deep learning framework is capable of identifying and quantifying NFT with a range of staining intensities and diverse morphologies. With our FCN model, we achieved high precision and recall in naive WSI semantic segmentation, correctly identifying tangle objects using a SegNet model trained for 200 epochs. Our FCN is efficient and well suited for the practical application of WSIs with average processing times of 45 min per WSI per GPU, enabling reliable and reproducible large-scale detection of tangles. We measured performance on test data of 50 pre-annotated regions on eight naive WSI across various tauopathies, resulting in the recall, precision, and an F1 score of 0.92, 0.72, and 0.81, respectively. Machine learning is a useful tool for complex pathological assessment of AD and other tauopathies. Using deep learning classifiers, we have the potential to integrate cell- and region-specific annotations with clinical, genetic, and molecular data, providing unbiased data for clinicopathological correlations that will enhance our knowledge of the neurodegeneration.
Collapse
Affiliation(s)
- Maxim Signaevsky
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marcel Prastawa
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Computational and Systems Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10025, USA
| | - Kurt Farrell
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nabil Tabish
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Elena Baldwin
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Natalia Han
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Megan A Iida
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - John Koll
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Computational and Systems Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10025, USA
| | - Clare Bryce
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dushyant Purohit
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vahram Haroutunian
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- J. James Peters VA Medical Center, Bronx, NY, USA
| | - Ann C McKee
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Pathology, Boston University School of Medicine, Boston, MA, 02118, USA
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, MA, 02118, USA
- Mental Illness Research, Education and Clinical Center, James J. Peters VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Veteran Affairs Medical Center, Bedford, MA, 01730, USA
| | - Thor D Stein
- Department of Pathology, Boston University School of Medicine, Boston, MA, 02118, USA
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, MA, 02118, USA
- Mental Illness Research, Education and Clinical Center, James J. Peters VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Veteran Affairs Medical Center, Bedford, MA, 01730, USA
| | - Charles L White
- Neuropathology Laboratory, Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jamie Walker
- Neuropathology Laboratory, Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Timothy E Richardson
- Neuropathology Laboratory, Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Russell Hanson
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michael J Donovan
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Computational and Systems Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10025, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Computational and Systems Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10025, USA
| | - Jack Zeineh
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Computational and Systems Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10025, USA
| | - Gerardo Fernandez
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Computational and Systems Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10025, USA
| | - John F Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
38
|
Scialò C, De Cecco E, Manganotti P, Legname G. Prion and Prion-Like Protein Strains: Deciphering the Molecular Basis of Heterogeneity in Neurodegeneration. Viruses 2019; 11:E261. [PMID: 30875755 PMCID: PMC6466326 DOI: 10.3390/v11030261] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/08/2019] [Accepted: 03/10/2019] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggests that neurodegenerative disorders share a common pathogenic feature: the presence of deposits of misfolded proteins with altered physicochemical properties in the Central Nervous System. Despite a lack of infectivity, experimental data show that the replication and propagation of neurodegenerative disease-related proteins including amyloid-β (Aβ), tau, α-synuclein and the transactive response DNA-binding protein of 43 kDa (TDP-43) share a similar pathological mechanism with prions. These observations have led to the terminology of "prion-like" to distinguish between conditions with noninfectious characteristics but similarities with the prion replication and propagation process. Prions are considered to adapt their conformation to changes in the context of the environment of replication. This process is known as either prion selection or adaptation, where a distinct conformer present in the initial prion population with higher propensity to propagate in the new environment is able to prevail over the others during the replication process. In the last years, many studies have shown that prion-like proteins share not only the prion replication paradigm but also the specific ability to aggregate in different conformations, i.e., strains, with relevant clinical, diagnostic and therapeutic implications. This review focuses on the molecular basis of the strain phenomenon in prion and prion-like proteins.
Collapse
Affiliation(s)
- Carlo Scialò
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), 34136 Trieste, Italy.
| | - Elena De Cecco
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), 34136 Trieste, Italy.
| | - Paolo Manganotti
- Clinical Unit of Neurology, Department of Medicine, Surgery and Health Sciences, University Hospital and Health Services of Trieste, University of Trieste, 34149 Trieste, Italy.
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), 34136 Trieste, Italy.
- ELETTRA Sincrotrone Trieste S.C.p.A, Basovizza, 34149 Trieste, Italy.
| |
Collapse
|
39
|
Chung DEC, Carlomagno Y, Cook CN, Jansen-West K, Daughrity L, Lewis-Tuffin LJ, Castanedes-Casey M, DeTure M, Dickson DW, Petrucelli L. Tau exhibits unique seeding properties in globular glial tauopathy. Acta Neuropathol Commun 2019; 7:36. [PMID: 30845985 PMCID: PMC6404306 DOI: 10.1186/s40478-019-0691-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
Tauopathies are neurodegenerative disorders characterized by aggregation of microtubule associated tau protein in neurons and glia. They are clinically and pathologically heterogeneous depending on the isoform of tau protein that accumulates (three or four 31-to-32-amino-acid repeats [3R or 4R] in the microtubule binding domain), as well as the cellular and neuroanatomical distribution of tau pathology. Growing evidence suggests that distinct tau conformers may contribute to the characteristic features of various tauopathies. Globular glial tauopathy (GGT) is a rare 4R tauopathy with globular cytoplasmic inclusions within neurons and glial cells. Given the unique cellular distribution and morphology of tau pathology in GGT, we sought to determine if tau species in GGT had distinctive biological properties. To address this question, we performed seeding analyses with postmortem brain tissues using a commercial tau biosensor cell line. We found that brain lysates from GGT cases had significantly higher seeding competency than other tauopathies, including corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), and Alzheimer’s disease (AD). The robust seeding activity of GGT brain lysates was independent of phosphorylated tau burden and diminished upon removal of tau from samples, suggesting that seeding properties were indeed mediated by tau in the lysates. In addition, cellular inclusions in the tau biosensor cell line induced by GGT had a distinct, globular morphology that was markedly different from inclusions induced by other tauopathies, further highlighting the unique nature of tau species in GGT. Characterization of different tau species in GGT showed that detergent-insoluble, fibril-like tau contained the highest seeding activity, as reflected in its ability to increase tau aggregation in primary glial cultures. Taken together, our data suggest that unique seeding properties differentiate GGT-tau from other tauopathies, which provides new insight into pathogenic heterogeneity of primary neurodegenerative tauopathies.
Collapse
|
40
|
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by extracellular β-amyloid plaques and intracellular neurofibrillary tangles (NFTs), which are considered as major targets for AD therapies. However, no effective therapy is available to cure or prevent the progression of AD up until now. Accumulation of NFTs, which consist of abnormally hyperphosphorylated tau, is directly correlated with the degree of dementia in AD patients. Emerging evidence indicates that the prion-like seeding and spreading of tau pathology may be the key driver of AD. In the past decades, greater understanding of tau pathway reveals new targets for the development of specific therapies. Here, we review the recent research progress in the mechanism underlying tau pathology in AD and briefly introduce tau-based therapeutics.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, PR China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York 10314, United States
| |
Collapse
|
41
|
Sadick JS, Liddelow SA. Don't forget astrocytes when targeting Alzheimer's disease. Br J Pharmacol 2019; 176:3585-3598. [PMID: 30636042 DOI: 10.1111/bph.14568] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
Astrocytes are essential for CNS health, regulating homeostasis, metabolism, and synaptic transmission. In addition to these and many other physiological roles, the pathological impact of astrocytes ("reactive astrocytes") in acute trauma and chronic disease like Alzheimer's disease (AD) is well established. Growing evidence supports a fundamental and active role of astrocytes in multiple neurodegenerative diseases. With a growing interest in normal astrocyte biology, and countless studies on changes in astrocyte function in the context of disease, it may be a surprise that no therapies exist incorporating astrocytes as key targets. Here, we examine unintentional effects of current AD therapies on astrocyte function and theorize how astrocytes may be intentionally targeted for more efficacious therapeutic outcomes. Given their integral role in normal neuronal functioning, incorporating astrocytes as key criteria for AD drug development can only lead to more effective therapies for the millions of AD sufferers worldwide. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Jessica S Sadick
- Neuroscience Institute, NYU Langone Medical Center, New York, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Langone Medical Center, New York, USA.,Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, USA.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
Fu H, Possenti A, Freer R, Nakano Y, Hernandez Villegas NC, Tang M, Cauhy PVM, Lassus BA, Chen S, Fowler SL, Figueroa HY, Huey ED, Johnson GVW, Vendruscolo M, Duff KE. A tau homeostasis signature is linked with the cellular and regional vulnerability of excitatory neurons to tau pathology. Nat Neurosci 2018; 22:47-56. [PMID: 30559469 PMCID: PMC6330709 DOI: 10.1038/s41593-018-0298-7] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/23/2018] [Indexed: 01/04/2023]
Abstract
Excitatory neurons are preferentially impaired in early Alzheimer's disease but the pathways contributing to their relative vulnerability remain largely unknown. Here we report that pathological tau accumulation takes place predominantly in excitatory neurons compared to inhibitory neurons, not only in the entorhinal cortex, a brain region affected in early Alzheimer's disease, but also in areas affected later by the disease. By analyzing RNA transcripts from single-nucleus RNA datasets, we identified a specific tau homeostasis signature of genes differentially expressed in excitatory compared to inhibitory neurons. One of the genes, BCL2-associated athanogene 3 (BAG3), a facilitator of autophagy, was identified as a hub, or master regulator, gene. We verified that reducing BAG3 levels in primary neurons exacerbated pathological tau accumulation, whereas BAG3 overexpression attenuated it. These results define a tau homeostasis signature that underlies the cellular and regional vulnerability of excitatory neurons to tau pathology.
Collapse
Affiliation(s)
- Hongjun Fu
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA.
| | - Andrea Possenti
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Rosie Freer
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Yoshikazu Nakano
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | | | - Maoping Tang
- Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Paula V M Cauhy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA.,Federal University of Uberlândia, Uberlândia, Brazil
| | - Benjamin A Lassus
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Shuo Chen
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Stephanie L Fowler
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Helen Y Figueroa
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Edward D Huey
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA.,Departments of Psychiatry and Neurology, Columbia University, New York, NY, USA
| | - Gail V W Johnson
- Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Karen E Duff
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
43
|
Luca A, Calandra C, Luca M. Molecular Bases of Alzheimer's Disease and Neurodegeneration: The Role of Neuroglia. Aging Dis 2018; 9:1134-1152. [PMID: 30574424 PMCID: PMC6284765 DOI: 10.14336/ad.2018.0201] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Neuroglia is an umbrella term indicating different cellular types that play a pivotal role in the brain, being involved in its development and functional homeostasis. Glial cells are becoming the focus of recent researches pertaining the pathogenesis of neurodegenerative disorders, Alzheimer's Disease (AD) in particular. In fact, activated microglia is the main determinant of neuroinflammation, contributing to neurodegeneration. In addition, the oxidative insult occurring during pathological brain aging can activate glial cells that, in turn, can favor the production of free radicals. Moreover, the recent Glycogen Synthase Kinase 3 (GSK-3) hypothesis of AD suggests that GSK3, involved in the regulation of glial cells functioning, could exert a role in amyloid deposition and tau hyper-phosphorylation. In this review, we briefly describe the main physiological functions of the glial cells and discuss the link between neuroglia and the most studied molecular bases of AD. In addition, we dedicate a section to the glial changes occurring in AD, with particular attention to their role in terms of neurodegeneration. In the light of the literature data, neuroglia could play a fundamental role in AD pathogenesis and progression. Further studies are needed to shed light on this topic.
Collapse
Affiliation(s)
- Antonina Luca
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| | - Carmela Calandra
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| | - Maria Luca
- Department of General Surgery and Medical-Surgical Specialties, Dermatology Clinic, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| |
Collapse
|
44
|
LoPresti P. Tau in Oligodendrocytes Takes Neurons in Sickness and in Health. Int J Mol Sci 2018; 19:ijms19082408. [PMID: 30111714 PMCID: PMC6121290 DOI: 10.3390/ijms19082408] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/31/2022] Open
Abstract
Oligodendrocytes (OLGs), the myelin-forming cells of the central nervous system (CNS), are lifelong partners of neurons. They adjust to the functional demands of neurons over the course of a lifetime to meet the functional needs of a healthy CNS. When this functional interplay breaks down, CNS degeneration follows. OLG processes are essential features for OLGs being able to connect with the neurons. As many as fifty cellular processes from a single OLG reach and wrap an equal number of axonal segments. The cellular processes extend to meet and wrap axonal segments with myelin. Further, transport regulation, which is critical for myelination, takes place within the cellular processes. Because the microtubule-associated protein tau plays a crucial role in cellular process extension and myelination, alterations of tau in OLGs have deleterious effects, resulting in neuronal malfunction and CNS degeneration. Here, we review current concepts on the lifelong role of OLGs and myelin for brain health and plasticity. We present key studies of tau in OLGs and select important studies of tau in neurons. The extensive work on tau in neurons has considerably advanced our understanding of how tau promotes either health or disease. Because OLGs are crucial to neuronal health at any age, an understanding of the functions and regulation of tau in OLGs could uncover new therapeutics for selective CNS neurodegenerative diseases.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, 1007 West Harrison Street, Chicago, IL 60607, USA.
| |
Collapse
|
45
|
Abstract
Astrocytes are involved in many diseases of the central nervous system, not only as reactive cells to neuronal damage but also as primary actors in the pathological process. Astrogliopathy is a term used to designate the involvement of astrocytes as key elements in the pathogenesis and pathology of diseases and injuries of the central nervous system. Astrocytopathy is utilized to name non-reactive astrogliosis covering hypertrophy, atrophy and astroglial degeneration with loss of function in astrocytes and pathological remodeling, as well as senescent changes. Astrogliopathy and astrocytopathy are hallmarks of tauopathies—neurodegenerative diseases with abnormal hyper-phosphorylated tau aggregates in neurons and glial cells. The involvement of astrocytes covers different disease-specific types such as tufted astrocytes, astrocytic plaques, thorn-shaped astrocytes, granular/fuzzy astrocytes, ramified astrocytes and astrocytes with globular inclusions, as well as others which are unnamed but not uncommon in familial frontotemporal degeneration linked to mutations in the tau gene. Knowledge of molecular differences among tau-containing astrocytes is only beginning, and their distinct functional implications remain rather poorly understood. However, tau-containing astrocytes in certain conditions have deleterious effects on neuronal function and nervous system integrity. Moreover, recent studies have shown that tau-containing astrocytes obtained from human brain tauopathies have a capacity for abnormal tau seeding and spreading in wild type mice. Inclusive conceptions include a complex scenario involving neurons, glial cells and local environmental factors that potentiate each other and promote disease progression in tauopathies.
Collapse
|
46
|
Muñoz-Mayorga D, Guerra-Araiza C, Torner L, Morales T. Tau Phosphorylation in Female Neurodegeneration: Role of Estrogens, Progesterone, and Prolactin. Front Endocrinol (Lausanne) 2018; 9:133. [PMID: 29643836 PMCID: PMC5882780 DOI: 10.3389/fendo.2018.00133] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
Sex differences are important to consider when studying different psychiatric, neurodevelopmental, and neurodegenerative disorders, including Alzheimer's disease (AD). These disorders can be affected by dimorphic changes in the central nervous system and be influenced by sex-specific hormones and neuroactive steroids. In fact, AD is more prevalent in women than in men. One of the main characteristics of AD is the formation of neurofibrillary tangles, composed of the phosphoprotein Tau, and neuronal loss in specific brain regions. The scope of this work is to review the existing evidence on how a set of hormones (estrogen, progesterone, and prolactin) affect tau phosphorylation in the brain of females under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Daniel Muñoz-Mayorga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
- *Correspondence: Teresa Morales,
| |
Collapse
|
47
|
Pierozan P, Biasibetti-Brendler H, Schmitz F, Ferreira F, Netto CA, Wyse ATS. Synergistic Toxicity of the Neurometabolites Quinolinic Acid and Homocysteine in Cortical Neurons and Astrocytes: Implications in Alzheimer's Disease. Neurotox Res 2017; 34:147-163. [PMID: 29124681 DOI: 10.1007/s12640-017-9834-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/22/2017] [Accepted: 10/18/2017] [Indexed: 12/19/2022]
Abstract
The brain of patients affected by Alzheimer's disease (AD) develops progressive neurodegeneration linked to the formation of proteins aggregates. However, their single actions cannot explain the extent of brain damage observed in this disorder, and the characterization of co-adjuvant involved in the early toxic processes evoked in AD is essential. In this line, quinolinic acid (QUIN) and homocysteine (Hcy) appear to be involved in the AD neuropathogenesis. Herein, we investigate the effects of QUIN and Hcy on early toxic events in cortical neurons and astrocytes. Exposure of primary cortical cultures to these neurometabolites for 24 h induced concentration-dependent neurotoxicity. In addition, QUIN (25 μM) and Hcy (30 μM) triggered ROS production, lipid peroxidation, diminished of Na+,K+-ATPase activity, and morphologic alterations, culminating in reduced neuronal viability by necrotic cell death. In astrocytes, QUIN (100 μM) and Hcy (30 μM) induced caspase-3-dependent apoptosis and morphologic alterations through oxidative status imbalance. To establish specific mechanisms, we preincubated cell cultures with different protective agents. The combined toxicity of QUIN and Hcy was attenuated by melatonin and Trolox in neurons and by NMDA antagonists and glutathione in astrocytes. Cellular death and morphologic alterations were prevented when co-culture was treated with metabolites, suggesting the activation of protector mechanisms dependent on soluble factors and astrocyte and neuron communication through gap junctions. These findings suggest that early damaging events involved in AD can be magnified by synergistic toxicity of the QUIN and Hcy. Therefore, this study opens new possibilities to elucidate the molecular mechanisms of neuron-astrocyte interactions and their role in neuroprotection against QUIN and Hcy.
Collapse
Affiliation(s)
- Paula Pierozan
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil.
| | - Helena Biasibetti-Brendler
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felipe Schmitz
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Ferreira
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Laboratório de Isquemia Cerebral e Psicobiologia dos Transtornos Mentais, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| | - Angela T S Wyse
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| |
Collapse
|
48
|
Kulbe JR, Hall ED. Chronic traumatic encephalopathy-integration of canonical traumatic brain injury secondary injury mechanisms with tau pathology. Prog Neurobiol 2017; 158:15-44. [PMID: 28851546 PMCID: PMC5671903 DOI: 10.1016/j.pneurobio.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
In recent years, a new neurodegenerative tauopathy labeled Chronic Traumatic Encephalopathy (CTE), has been identified that is believed to be primarily a sequela of repeated mild traumatic brain injury (TBI), often referred to as concussion, that occurs in athletes participating in contact sports (e.g. boxing, American football, Australian football, rugby, soccer, ice hockey) or in military combatants, especially after blast-induced injuries. Since the identification of CTE, and its neuropathological finding of deposits of hyperphosphorylated tau protein, mechanistic attention has been on lumping the disorder together with various other non-traumatic neurodegenerative tauopathies. Indeed, brains from suspected CTE cases that have come to autopsy have been confirmed to have deposits of hyperphosphorylated tau in locations that make its anatomical distribution distinct for other tauopathies. The fact that these individuals experienced repetitive TBI episodes during their athletic or military careers suggests that the secondary injury mechanisms that have been extensively characterized in acute TBI preclinical models, and in TBI patients, including glutamate excitotoxicity, intracellular calcium overload, mitochondrial dysfunction, free radical-induced oxidative damage and neuroinflammation, may contribute to the brain damage associated with CTE. Thus, the current review begins with an in depth analysis of what is known about the tau protein and its functions and dysfunctions followed by a discussion of the major TBI secondary injury mechanisms, and how the latter have been shown to contribute to tau pathology. The value of this review is that it might lead to improved neuroprotective strategies for either prophylactically attenuating the development of CTE or slowing its progression.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States
| | - Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States.
| |
Collapse
|
49
|
Zhou Y, Bai B. Tau and Pet/Mri Imaging Biomarkers for Detecting and Diagnosing Early Dementia. JACOBS JOURNAL OF MEDICAL DIAGNOSIS AND MEDICAL IMAGING 2017; 2:017. [PMID: 29026896 PMCID: PMC5634528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Specific objective of this project is to quantify the neuropathological tau depositions in brain regions and to investigate primary age-related tau pathology and associations with amyloid and glucose-metabolism, neurocognitive tests and MRI metrics. Preliminary results demonstrated higher temporal deposition especially in the hippocampus and the inferior temporal regions compared to mean cortical deposition. Quantitative Braak stage-based regional analyses found highest tau deposition in the Braak stage II in the preclinical samples. There was significant age related higher tau deposition in temporal cortex, while non-significant correlation in mean cortical region. Significant correlations were found between tau deposition and memory as well as executive function scores, especially from middle and inferior temporal cortex; and significant correlations between tau and MRI metrics including diffusion, perfusion, functional and structural connectivities. Our results confirm current notion of this new tau tracer for reliably and consistently labeling and quantifying in vivo human taupathy in early dementia.
Collapse
Affiliation(s)
- Yongxia Zhou
- Department of Radiology/Center for Biomedical Imaging, NYU Langone Medical Center, New York, NY
| | - Bing Bai
- Department of Radiology, University of Southern California, Los Angeles, CA
| |
Collapse
|
50
|
Kneynsberg A, Combs B, Christensen K, Morfini G, Kanaan NM. Axonal Degeneration in Tauopathies: Disease Relevance and Underlying Mechanisms. Front Neurosci 2017; 11:572. [PMID: 29089864 PMCID: PMC5651019 DOI: 10.3389/fnins.2017.00572] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/29/2017] [Indexed: 12/14/2022] Open
Abstract
Tauopathies are a diverse group of diseases featuring progressive dying-back neurodegeneration of specific neuronal populations in association with accumulation of abnormal forms of the microtubule-associated protein tau. It is well-established that the clinical symptoms characteristic of tauopathies correlate with deficits in synaptic function and neuritic connectivity early in the course of disease, but mechanisms underlying these critical pathogenic events are not fully understood. Biochemical in vitro evidence fueled the widespread notion that microtubule stabilization represents tau's primary biological role and that the marked atrophy of neurites observed in tauopathies results from loss of microtubule stability. However, this notion contrasts with the mild phenotype associated with tau deletion. Instead, an analysis of cellular hallmarks common to different tauopathies, including aberrant patterns of protein phosphorylation and early degeneration of axons, suggests that alterations in kinase-based signaling pathways and deficits in axonal transport (AT) associated with such alterations contribute to the loss of neuronal connectivity triggered by pathogenic forms of tau. Here, we review a body of literature providing evidence that axonal pathology represents an early and common pathogenic event among human tauopathies. Observations of axonal degeneration in animal models of specific tauopathies are discussed and similarities to human disease highlighted. Finally, we discuss potential mechanistic pathways other than microtubule destabilization by which disease-related forms of tau may promote axonopathy.
Collapse
Affiliation(s)
- Andrew Kneynsberg
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Benjamin Combs
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Kyle Christensen
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Nicholas M Kanaan
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|