1
|
Shwab EK, Man Z, Gingerich DC, Gamache J, Garrett ME, Serrano GE, Beach TG, Crawford GE, Ashley‐Koch AE, Chiba‐Falek O. Comparative mapping of single-cell transcriptomic landscapes in neurodegenerative diseases. Alzheimers Dement 2025; 21:e70012. [PMID: 40344336 PMCID: PMC12061851 DOI: 10.1002/alz.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and Parkinson's disease (PD) represent a spectrum of neurodegenerative diseases (NDDs). Here, we performed the first direct comparison of their transcriptomic landscapes. METHODS We profiled whole transcriptomes of NDD cortical tissue by single-nucleus RNA sequencing, using computational analyses to identify common and distinct differentially expressed genes (DEGs), pathways, vulnerable and disease-driver cell subtypes, and altered cell-to-cell interactions. RESULTS The same inhibitory neuron subtype was depleted in both AD and DLB. Potentially disease-driving neuronal cell subtypes were identified in both PD and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. DEGs were most commonly shared across NDDs within inhibitory neuron subtypes. Overall, AD and PD showed greatest transcriptomic divergence, while DLB exhibited an intermediate signature. DISCUSSION These results may help explain the clinicopathological spectrum of these NDDs and provide unique insights into shared and distinct molecular mechanisms underlying pathogenesis. HIGHLIGHTS The same vulnerable inhibitory neuron subtype population was depleted in both Alzheimer's disease (AD) and dementia with Lewy bodies (DLB). Potentially disease-driving neuronal cell subtypes were discovered in both Parkinson's disease (PD) and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. Differentially expressed genes were most commonly shared across neurodegenerative diseases in inhibitory neuron types. AD and PD had the greatest transcriptomic divergence, with DLB showing an intermediate signature.
Collapse
Affiliation(s)
- E. Keats Shwab
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Zhaohui Man
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Daniel C. Gingerich
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Julia Gamache
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Melanie E. Garrett
- Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNorth CarolinaUSA
| | | | | | - Gregory E. Crawford
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of PediatricsDivision of Medical GeneticsDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Advanced Genomic TechnologiesDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Allison E. Ashley‐Koch
- Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of MedicineDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ornit Chiba‐Falek
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| |
Collapse
|
2
|
Fischer AL, Schmitz M, Thom T, Zafar S, Younas N, da Silva Correia S, da Silva Correia A, Eyyuboglu SC, Zerr I. Alpha-Synuclein Demonstrates Varying Binding Affinities With Different Tau Isoforms. J Neurochem 2025; 169:e70053. [PMID: 40165586 DOI: 10.1111/jnc.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
The hallmark of various neurodegenerative diseases is the accumulation and aggregation of amyloidogenic proteins, such as amyloid-beta (Aβ) and tau in Alzheimer's disease (AD) and alpha-synuclein (aSyn) in synucleinopathies. Many neurodegenerative diseases express mixed pathology. For instance, Lewy bodies are reported in tauopathies and neurofibrillary tau-tangles are detected in synucleinopathies, suggesting a potential co-existence or crosstalk of misfolded aSyn and tau. In the present study, we investigated the binding characteristics of monomeric aSyn with different tau isoforms by using surface plasmon resonance (SPR) spectroscopy allowing monitoring direct protein-protein interactions and their potential co-localization using SH-SY5Y cells. The calculation of the binding parameters (association and dissociation rate constants) indicated the strongest binding affinity between aSyn and tau isoform 1N3R followed by tau 2N3R and tau 2N4R. Co-localization studies in SH-SY5Y cells, treated with aSyn and all six tau isoforms revealed an intracellular co-localization of aSyn with different isoforms of tau. Subcellular fractionation confirmed the cellular uptake and colocalization of tau and aSyn in the same compartment, showing their expression in membrane, nuclear, and cytoskeletal fractions. Understanding the intricate interplay between aSyn and tau is crucial for unraveling the pathophysiology of PD, AD, and related neurodegenerative disorders, ultimately paving the way for the development of effective treatments targeting this interaction. In conclusion, our data indicate that aSyn and tau are direct interaction partners with varying binding affinities depending on the tau isoform. This interaction may be significant for understanding the pathophysiology of dementia with mixed pathologies.
Collapse
Affiliation(s)
- Anna-Lisa Fischer
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Tobias Thom
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Saima Zafar
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Neelam Younas
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Susana da Silva Correia
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Angela da Silva Correia
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Sezgi Canaslan Eyyuboglu
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
3
|
Franzmeier N, Roemer-Cassiano SN, Bernhardt AM, Dehsarvi A, Dewenter A, Steward A, Biel D, Frontzkowski L, Zhu Z, Gnörich J, Pescoller J, Wagner F, Hirsch F, de Bruin H, Ossenkoppele R, Palleis C, Strübing F, Schöll M, Levin J, Brendel M, Höglinger GU. Alpha synuclein co-pathology is associated with accelerated amyloid-driven tau accumulation in Alzheimer's disease. Mol Neurodegener 2025; 20:31. [PMID: 40098057 PMCID: PMC11916967 DOI: 10.1186/s13024-025-00822-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/02/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Aggregated alpha-Synuclein (αSyn) is a hallmark pathology in Parkinson's disease but also one of the most common co-pathologies in Alzheimer's disease (AD). Preclinical studies suggest that αSyn can exacerbate tau aggregation, implying that αSyn co-pathology may specifically contribute to the Aβ-induced aggregation of tau that drives neurodegeneration and cognitive decline in AD. To investigate this, we combined a novel CSF-based seed-amplification assay (SAA) to determine αSyn positivity with amyloid- and tau-PET neuroimaging in a large cohort ranging from cognitively normal individuals to those with dementia, examining whether αSyn co-pathology accelerates Aβ-driven tau accumulation and cognitive decline. METHODS In 284 Aβ-positive and 308 Aβ-negative subjects, we employed amyloid-PET, Flortaucipir tau-PET, and a CSF-based αSyn seed-amplification assay (SAA) to detect in vivo αSyn aggregation. CSF p-tau181 measures were available for 384 subjects to assess earliest tau abnormalities. A subset of 155 Aβ-positive and 135 Aβ-negative subjects underwent longitudinal tau-PET over approximately 2.5 years. Using linear regression models, we analyzed whether αSyn SAA positivity was linked to stronger Aβ-related increases in baseline fluid and PET tau biomarkers, faster Aβ-driven tau-PET increase, and more rapid cognitive decline. RESULTS αSyn SAA positivity was more common in Aβ + vs. Aβ- subjects and increased with clinical severity (p < 0.001). Most importantly, αSyn positivity was also associated with greater amyloid-associated CSF p-tau181 increases (p = 0.005) and higher tau-PET levels in AD-typical brain regions (p = 0.006). Longitudinal analyses confirmed further that αSyn positivity was associated with faster amyloid-related tau accumulation (p = 0.029) and accelerated amyloid-related cognitive decline, potentially driven driven by stronger tau pathology. CONCLUSIONS Our findings suggest that αSyn co-pathology, detectable via CSF-based SAAs, is more prevalent in advanced AD and contributes to the development of aggregated tau pathology thereby driving faster cognitive decline. This highlights that a-Syn co-pathology may specifically accelerate amyloid-driven tau pathophysiology in AD, underscoring the need to consider αSyn in AD research and treatment strategies.
Collapse
Affiliation(s)
- Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal and Gothenburg, Sweden.
| | - Sebastian Niclas Roemer-Cassiano
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Neurology, LMU University Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Maximilian Bernhardt
- Department of Neurology, LMU University Hospital, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Lukas Frontzkowski
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Zeyu Zhu
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Julia Pescoller
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Fabian Wagner
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Fabian Hirsch
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Hannah de Bruin
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Carla Palleis
- Department of Neurology, LMU University Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Felix Strübing
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany
| | - Michael Schöll
- The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal and Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
- Department of Neuropsychiatry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johannes Levin
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Günter U Höglinger
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
4
|
Lucas L, Tsoi PS, Quan MD, Choi KJ, Ferreon JC, Ferreon ACM. Tubulin transforms Tau and α-synuclein condensates from pathological to physiological. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640500. [PMID: 40060635 PMCID: PMC11888465 DOI: 10.1101/2025.02.27.640500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Proteins phase-separate to form condensates that partition and concentrate biomolecules into membraneless compartments. These condensates can exhibit dichotomous behaviors in biology by supporting cellular physiology or instigating pathological protein aggregation 1-3 . Tau and α- synuclein (αSyn) are neuronal proteins that form heterotypic (Tau:αSyn) condensates associated with both physiological and pathological processes. Tau and αSyn functionally regulate microtubules 8-12 , but are also known to misfold and co-deposit in aggregates linked to various neurodegenerative diseases 4,5,6,7 , which highlights the paradoxically ambivalent effect of Tau:αSyn condensation in health and disease. Here, we show that tubulin modulates Tau:αSyn condensates by promoting microtubule interactions, competitively inhibiting the formation of homotypic and heterotypic pathological oligomers. In the absence of tubulin, Tau-driven protein condensation accelerates the formation of toxic Tau:αSyn heterodimers and amyloid fibrils. However, tubulin partitioning into Tau:αSyn condensates modulates protein interactions, promotes microtubule polymerization, and prevents Tau and αSyn oligomerization and aggregation. We distinguished distinct Tau and αSyn structural states adopted in tubulin-absent (pathological) and tubulin-rich (physiological) condensates, correlating compact conformations with aggregation and extended conformations with function. Furthermore, using various neuronal cell models, we showed that loss of stable microtubules, which occurs in Alzheimer's disease and Parkinsons disease patients 13,14 , results in pathological oligomer formation and loss of neurites, and that functional condensation using an inducible optogenetic Tau construct resulted in microtubule stablization. Our results identify that tubulin is a critical modulator in switching Tau:αSyn pathological condensates to physiological, mechanistically relating the loss of stable microtubules with disease progression. Tubulin restoration strategies and Tau-mediated microtubule stabilization can be potential therapies targeting both Tau-specific and Tau/αSyn mixed pathologies.
Collapse
|
5
|
Fischer DL, Menard M, Abdelaziz OZ, Kanaan NM, Cobbs VG, Kennedy RE, Serrano GE, Beach TG, Volpicelli-Daley LA. Distinct subcellular localization of tau and alpha-synuclein in lewy body disease. Acta Neuropathol Commun 2025; 13:14. [PMID: 39838428 PMCID: PMC11752823 DOI: 10.1186/s40478-024-01913-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Lewy bodies and neurofibrillary tangles, composed of α-synuclein (α-syn) and tau, respectively, often are found together in the same brain and correlate with worsening cognition. Human postmortem studies show colocalization of α-syn and tau occurs in Lewy bodies, but with limited effort to quantify colocalization. In this study, postmortem middle temporal gyrus tissue from decedents (n = 9) without temporal lobe disease (control) or with Lewy body disease (LBD) was immunofluorescently labeled with antibodies to phosphorylated α-syn (p-α-syn), tau phosphorylated at Ser202/Thr205 (p-tau), or exposure of tau's phosphatase-activating domain (PAD-tau) as a marker of early tau aggregates. Immunofluorescence for major-histocompatibility complex class 2 (MHCII) and ionized calcium binding adaptor molecule 1 (Iba1) also was performed because inflammation is an additional pathological hallmark of LBDs, and they were a positive control for two markers known to colocalize. The abundance of p-α-syn, p-tau, and MHCII was significantly associated with diagnosis of LBD. Quantification of colocalization showed that MHCII and Iba1 colocalized, demonstrating activated immune cells are mostly microglia. However, p-α-syn rarely colocalized with p-tau or PAD-tau, although the overlap of p-α-syn with PAD-tau was significantly associated with LBD. In the rare cases pathologic α-syn and pathologic tau were found in the same Lewy body or Lewy neurite, tau appeared to surround α-syn but did not colocalize within the same structure. The relationship between tau and α-syn copathology is important for explaining clinical symptoms, severity, and progression, but there is no evidence for frequent, direct protein-protein interactions in the middle temporal gyrus.
Collapse
Affiliation(s)
- D Luke Fischer
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Marissa Menard
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Omar Z Abdelaziz
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Virginia G Cobbs
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard E Kennedy
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
6
|
Shwab EK, Man Z, Gingerich DC, Gamache J, Garrett ME, Serrano GE, Beach TG, Crawford GE, Ashley-Koch AE, Chiba-Falek O. Comparative mapping of single-cell transcriptomic landscapes in neurodegenerative diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628436. [PMID: 39764045 PMCID: PMC11702568 DOI: 10.1101/2024.12.13.628436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), Dementia with Lewy bodies (DLB), and Parkinson's disease (PD) represent a spectrum of neurodegenerative disorders (NDDs). Here, we performed the first direct comparison of their transcriptomic landscapes. METHODS We profiled the whole transcriptomes of NDD cortical tissue by snRNA-seq. We used computational analyses to identify common and distinct differentially expressed genes (DEGs), biological pathways, vulnerable and disease-driver cell subtypes, and alteration in cell-to-cell interactions. RESULTS The same vulnerable inhibitory neuron subtype was depleted in both AD and DLB. Potentially disease-driving neuronal cell subtypes were present in both PD and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. DEGs were most commonly shared across NDDs within inhibitory neuron subtypes. Overall, we observed the greatest transcriptomic divergence between AD and PD, while DLB exhibited an intermediate transcriptomic signature. DISCUSSION These results help explain the clinicopathological spectrum of this group of NDDs and provide unique insights into the shared and distinct molecular mechanisms underlying the pathogenesis of NDDs.
Collapse
Affiliation(s)
- E. Keats Shwab
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Zhaohui Man
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Daniel C. Gingerich
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Melanie E. Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA
| | - Geidy E. Serrano
- Banner Sun Health Research Institute, Sun City, Arizona, 85351, USA
| | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, Arizona, 85351, USA
| | - Gregory E. Crawford
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, NC, 27708, USA
- Center for Advanced Genomic Technologies, Duke University Medical Center, Durham, NC, 27708, USA
| | - Allison E. Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| |
Collapse
|
7
|
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
|
8
|
Langerscheidt F, Wied T, Al Kabbani MA, van Eimeren T, Wunderlich G, Zempel H. Genetic forms of tauopathies: inherited causes and implications of Alzheimer's disease-like TAU pathology in primary and secondary tauopathies. J Neurol 2024; 271:2992-3018. [PMID: 38554150 PMCID: PMC11136742 DOI: 10.1007/s00415-024-12314-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024]
Abstract
Tauopathies are a heterogeneous group of neurologic diseases characterized by pathological axodendritic distribution, ectopic expression, and/or phosphorylation and aggregation of the microtubule-associated protein TAU, encoded by the gene MAPT. Neuronal dysfunction, dementia, and neurodegeneration are common features of these often detrimental diseases. A neurodegenerative disease is considered a primary tauopathy when MAPT mutations/haplotypes are its primary cause and/or TAU is the main pathological feature. In case TAU pathology is observed but superimposed by another pathological hallmark, the condition is classified as a secondary tauopathy. In some tauopathies (e.g. MAPT-associated frontotemporal dementia (FTD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Alzheimer's disease (AD)) TAU is recognized as a significant pathogenic driver of the disease. In many secondary tauopathies, including Parkinson's disease (PD) and Huntington's disease (HD), TAU is suggested to contribute to the development of dementia, but in others (e.g. Niemann-Pick disease (NPC)) TAU may only be a bystander. The genetic and pathological mechanisms underlying TAU pathology are often not fully understood. In this review, the genetic predispositions and variants associated with both primary and secondary tauopathies are examined in detail, assessing evidence for the role of TAU in these conditions. We highlight less common genetic forms of tauopathies to increase awareness for these disorders and the involvement of TAU in their pathology. This approach not only contributes to a deeper understanding of these conditions but may also lay the groundwork for potential TAU-based therapeutic interventions for various tauopathies.
Collapse
Affiliation(s)
- Felix Langerscheidt
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Tamara Wied
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Von-Liebig-Str. 20, 53359, Rheinbach, Germany
| | - Mohamed Aghyad Al Kabbani
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Thilo van Eimeren
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
| | - Gilbert Wunderlich
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
- Center for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Hans Zempel
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
9
|
Jin M, Wang S, Gao X, Zou Z, Hirotsune S, Sun L. Pathological and physiological functional cross-talks of α-synuclein and tau in the central nervous system. Neural Regen Res 2024; 19:855-862. [PMID: 37843221 PMCID: PMC10664117 DOI: 10.4103/1673-5374.382231] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/05/2023] [Accepted: 07/12/2023] [Indexed: 10/17/2023] Open
Abstract
α-Synuclein and tau are abundant multifunctional brain proteins that are mainly expressed in the presynaptic and axonal compartments of neurons, respectively. Previous works have revealed that intracellular deposition of α-synuclein and/or tau causes many neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. Despite intense investigation, the normal physiological functions and roles of α-synuclein and tau are still unclear, owing to the fact that mice with knockout of either of these proteins do not present apparent phenotypes. Interestingly, the co-occurrence of α-synuclein and tau aggregates was found in post-mortem brains with synucleinopathies and tauopathies, some of which share similarities in clinical manifestations. Furthermore, the direct interaction of α-synuclein with tau is considered to promote the fibrillization of each of the proteins in vitro and in vivo. On the other hand, our recent findings have revealed that α-synuclein and tau are cooperatively involved in brain development in a stage-dependent manner. These findings indicate strong cross-talk between the two proteins in physiology and pathology. In this review, we provide a summary of the recent findings on the functional roles of α-synuclein and tau in the physiological conditions and pathogenesis of neurodegenerative diseases. A deep understanding of the interplay between α-synuclein and tau in physiological and pathological conditions might provide novel targets for clinical diagnosis and therapeutic strategies to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingyue Jin
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Shengming Wang
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Xiaodie Gao
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Zhenyou Zou
- Department of Scientific Research, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou, Guangxi Zhuang Autonomous Region, China
| | - Shinji Hirotsune
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Liyuan Sun
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
10
|
Li W, Li JY. Overlaps and divergences between tauopathies and synucleinopathies: a duet of neurodegeneration. Transl Neurodegener 2024; 13:16. [PMID: 38528629 DOI: 10.1186/s40035-024-00407-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/28/2024] [Indexed: 03/27/2024] Open
Abstract
Proteinopathy, defined as the abnormal accumulation of proteins that eventually leads to cell death, is one of the most significant pathological features of neurodegenerative diseases. Tauopathies, represented by Alzheimer's disease (AD), and synucleinopathies, represented by Parkinson's disease (PD), show similarities in multiple aspects. AD manifests extrapyramidal symptoms while dementia is also a major sign of advanced PD. We and other researchers have sequentially shown the cross-seeding phenomenon of α-synuclein (α-syn) and tau, reinforcing pathologies between synucleinopathies and tauopathies. The highly overlapping clinical and pathological features imply shared pathogenic mechanisms between the two groups of disease. The diagnostic and therapeutic strategies seemingly appropriate for one distinct neurodegenerative disease may also apply to a broader spectrum. Therefore, a clear understanding of the overlaps and divergences between tauopathy and synucleinopathy is critical for unraveling the nature of the complicated associations among neurodegenerative diseases. In this review, we discuss the shared and diverse characteristics of tauopathies and synucleinopathies from aspects of genetic causes, clinical manifestations, pathological progression and potential common therapeutic approaches targeting the pathology, in the aim to provide a timely update for setting the scheme of disease classification and provide novel insights into the therapeutic development for neurodegenerative diseases.
Collapse
Affiliation(s)
- Wen Li
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, China Medical University, Shenyang, 110122, China
| | - Jia-Yi Li
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, China Medical University, Shenyang, 110122, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, BMC A10, 22184, Lund, Sweden.
| |
Collapse
|
11
|
Walker L, Attems J. Prevalence of Concomitant Pathologies in Parkinson's Disease: Implications for Prognosis, Diagnosis, and Insights into Common Pathogenic Mechanisms. JOURNAL OF PARKINSON'S DISEASE 2024; 14:35-52. [PMID: 38143370 PMCID: PMC10836576 DOI: 10.3233/jpd-230154] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/11/2023] [Indexed: 12/26/2023]
Abstract
Pathologies characteristic of Alzheimer's disease (i.e., hyperphosphorylated tau and amyloid-β (Aβ) plaques), cardiovascular disease, and limbic predominant TDP-43 encephalopathy (LATE) often co-exist in patients with Parkinson's disease (PD), in addition to Lewy body pathology (α-synuclein). Numerous studies point to a putative synergistic relationship between hyperphosphorylation tau, Aβ, cardiovascular lesions, and TDP-43 with α-synuclein, which may alter the stereotypical pattern of pathological progression and accelerate cognitive decline. Here we discuss the prevalence and relationships between common concomitant pathologies observed in PD. In addition, we highlight shared genetic risk factors and developing biomarkers that may provide better diagnostic accuracy for patients with PD that have co-existing pathologies. The tremendous heterogeneity observed across the PD spectrum is most likely caused by the complex interplay between pathogenic, genetic, and environmental factors, and increasing our understanding of how these relate to idiopathic PD will drive research into finding accurate diagnostic tools and disease modifying therapies.
Collapse
Affiliation(s)
- Lauren Walker
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Johannes Attems
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
12
|
Chen K, Tang F, Du B, Yue Z, Jiao L, Ding X, Tuo Q, Meng J, He S, Dai L, Lei P, Wei X. Leucine-rich repeat kinase 2 (LRRK2) inhibition upregulates microtubule-associated protein 1B to ameliorate lysosomal dysfunction and parkinsonism. MedComm (Beijing) 2023; 4:e429. [PMID: 38020716 PMCID: PMC10661827 DOI: 10.1002/mco2.429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
Mutations in LRRK2 (encoding leucine-rich repeat kinase 2 protein, LRRK2) are the most common genetic risk factors for Parkinson's disease (PD), and increased LRRK2 kinase activity was observed in sporadic PD. Therefore, inhibition of LRRK2 has been tested as a disease-modifying therapeutic strategy using the LRRK2 mutant mice and sporadic PD. Here, we report a newly designed molecule, FL090, as a LRRK2 kinase inhibitor, verified in cell culture and animal models of PD. Using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice and SNCA A53T transgenic mice, FL090 ameliorated motor dysfunctions, reduced LRRK2 kinase activity, and rescued loss in the dopaminergic neurons in the substantia nigra. Notably, by RNA-Seq analysis, we identified microtubule-associated protein 1 (MAP1B) as a crucial mediator of FL090's neuroprotective effects and found that MAP1B and LRRK2 co-localize. Overexpression of MAP1B rescued 1-methyl-4-phenylpyridinium induced cytotoxicity through rescuing the lysosomal function, and the protective effect of FL090 was lost in MAP1B knockout cells. Further studies may be focused on the in vivo mechanisms of MAP1B and microtubule function in PD. Collectively, these findings highlight the potential of FL090 as a therapeutic agent for sporadic PD and familial PD without LRRK2 mutations.
Collapse
Affiliation(s)
- Kang Chen
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Fei Tang
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Bin Du
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Zhe‐Zhou Yue
- Guizhou Yiluoqini Techno. Co., Ltd, Guizhou Shuanglong Airport Economic ZoneGuiyangP. R. China
| | - Ling‐Ling Jiao
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Xu‐Long Ding
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Qing‐Zhang Tuo
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Jie Meng
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Si‐Yu He
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Lunzhi Dai
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
| | - Xia‐Wei Wei
- Department of Neurology and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan University, and Collaborative Center for BiotherapyChengduP. R. China
- Guizhou Yiluoqini Techno. Co., Ltd, Guizhou Shuanglong Airport Economic ZoneGuiyangP. R. China
| |
Collapse
|
13
|
Amirian R, Badrbani MA, Derakhshankhah H, Izadi Z, Shahbazi MA. Targeted protein degradation for the treatment of Parkinson's disease: Advances and future perspective. Biomed Pharmacother 2023; 166:115408. [PMID: 37651798 DOI: 10.1016/j.biopha.2023.115408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023] Open
Abstract
Parkinson's disease (PD) is a progressive disorder that belongs to a class of neurodegenerative disorders (NDs) called Synucleinopathies. It has characterized by the misfolding and aggregation of a-synuclein. Our understanding of PD continues to evolve, and so does our approach to treatment. including therapies aimed at delaying pathology, quitting neuronal loss, and shortening the course of the disease by selectively targeting essential proteins suspected to play a role in PD pathogenesis. One emerging approach that is generating significant interest is Targeted Protein Degradation (TPD). TPD is an innovative method that allows us to specifically break down certain proteins using specially designed molecules or peptides, like PROteolysis-TArgeting-Chimera (PROTACs). This approach holds great promise, particularly in the context of NDs. In this review, we will briefly explain PD and its pathogenesis, followed by discussing protein degradation systems and TPD strategy in PD by reviewing synthesized small molecules and peptides. Finally, future perspectives and challenges in the field are discussed.
Collapse
Affiliation(s)
- Roshanak Amirian
- Student research committee, School of pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Azadi Badrbani
- Student research committee, School of pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Derakhshankhah
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zhila Izadi
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
14
|
Kulichikhin KY, Malikova OA, Zobnina AE, Zalutskaya NM, Rubel AA. Interaction of Proteins Involved in Neuronal Proteinopathies. Life (Basel) 2023; 13:1954. [PMID: 37895336 PMCID: PMC10608209 DOI: 10.3390/life13101954] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/04/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Proteinopathy is characterized by the accumulation of aggregates of a specific protein in a target organ, tissue, or cell. The aggregation of the same protein can cause different pathologies as single protein can adopt various amyloidogenic, disease-specific conformations. The conformation governs the interaction of amyloid aggregates with other proteins that are prone to misfolding and, thus, determines disease-specific spectrum of concomitant pathologies. In this regard, a detailed description of amyloid protein conformation as well as spectrum of its interaction with other proteins become a key point for drafting of precise description of the disease. The majority of clinical cases of neuronal proteinopathies is caused by the aggregation of rather limited range of amyloidogenic proteins. Here, we provided the characterization of pathologies, related to the aggregation of amyloid β peptide, tau protein, α-synuclein, TDP-43, and amylin, giving a short description of pathologies themselves, recent advances in elucidation of misfolded protein conformation, with emphasis on those protein aggregates extracted from biological samples, what is known about the interaction of this proteins, and the influence of this interaction on the progression of underlying disease and comorbidities.
Collapse
Affiliation(s)
- Konstantin Y. Kulichikhin
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| | - Oksana A. Malikova
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| | - Anastasia E. Zobnina
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| | - Natalia M. Zalutskaya
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 192019 St. Petersburg, Russia;
| | - Aleksandr A. Rubel
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| |
Collapse
|
15
|
The central role of tau in Alzheimer’s disease: From neurofibrillary tangle maturation to the induction of cell death. Brain Res Bull 2022; 190:204-217. [DOI: 10.1016/j.brainresbull.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
|
16
|
Pan L, Meng L, He M, Zhang Z. Tau in the Pathophysiology of Parkinson's Disease. J Mol Neurosci 2021; 71:2179-2191. [PMID: 33459970 PMCID: PMC8585831 DOI: 10.1007/s12031-020-01776-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
The pathological hallmarks of Parkinson's disease (PD) are the progressive loss of dopaminergic neurons in the substantia nigra and the formation of Lewy bodies (LBs) in remaining neurons. LBs primarily consist of aggregated α-Synuclein (α-Syn). However, accumulating evidence suggests that Tau, which is associated with tauopathies such as Alzheimer's disease (AD), progressive supranuclear palsy (PSP), and argyrophilic grain disease, is also involved in the pathophysiology of PD. A genome-wide association study (GWAS) identified MAPT, the gene encoding the Tau protein, as a risk gene for PD. Autopsy of PD patients also revealed the colocalization of Tau and α-Syn in LBs. Experimental evidence has shown that Tau interacts with α-Syn and influences the pathology of α-Syn in PD. In this review, we discuss the structure and function of Tau and provide a summary of the current evidence supporting Tau's involvement as either an active or passive element in the pathophysiology of PD, which may provide novel targets for the early diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Lina Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Mingyang He
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
17
|
Mroczek K, Fernando S, Fisher PR, Annesley SJ. Interactions and Cytotoxicity of Human Neurodegeneration- Associated Proteins Tau and α-Synuclein in the Simple Model Dictyostelium discoideum. Front Cell Dev Biol 2021; 9:741662. [PMID: 34552934 PMCID: PMC8450459 DOI: 10.3389/fcell.2021.741662] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/19/2021] [Indexed: 11/17/2022] Open
Abstract
The abnormal accumulation of the tau protein into aggregates is a hallmark in neurodegenerative diseases collectively known as tauopathies. In normal conditions, tau binds off and on microtubules aiding in their assembly and stability dependent on the phosphorylation state of the protein. In disease-affected neurons, hyperphosphorylation leads to the accumulation of the tau protein into aggregates, mainly neurofibrillary tangles (NFT) which have been seen to colocalise with other protein aggregates in neurodegeneration. One such protein is α-synuclein, the main constituent of Lewy bodies (LB), a hallmark of Parkinson's disease (PD). In many neurodegenerative diseases, including PD, the colocalisation of tau and α-synuclein has been observed, suggesting possible interactions between the two proteins. To explore the cytotoxicity and interactions between these two proteins, we expressed full length human tau and α-synuclein in Dictyostelium discoideum alone, and in combination. We show that tau is phosphorylated in D. discoideum and colocalises closely (within 40 nm) with tubulin throughout the cytoplasm of the cell as well as with α-synuclein at the cortex. Expressing wild type α-synuclein alone caused inhibited growth on bacterial lawns, phagocytosis and intracellular Legionella proliferation rates, but activated mitochondrial respiration and non-mitochondrial oxygen consumption. The expression of tau alone impaired multicellular morphogenesis, axenic growth and phototaxis, while enhancing intracellular Legionella proliferation. Direct respirometric assays showed that tau impairs mitochondrial ATP synthesis and increased the "proton leak," while having no impact on respiratory complex I or II function. In most cases depending on the phenotype, the coexpression of tau and α-synuclein exacerbated (phototaxis, fruiting body morphology), or reversed (phagocytosis, growth on plates, mitochondrial respiratory function, Legionella proliferation) the defects caused by either tau or α-synuclein expressed individually. Proteomics data revealed distinct patterns of dysregulation in strains ectopically expressing tau or α-synuclein or both, but down regulation of expression of cytoskeletal proteins was apparent in all three groups and most evident in the strain expressing both proteins. These results indicate that tau and α-synuclein exhibit different but overlapping patterns of intracellular localisation, that they individually exert distinct but overlapping patterns of cytotoxic effects and that they interact, probably physically in the cell cortex as well as directly or indirectly in affecting some phenotypes. The results show the efficacy of using D. discoideum as a model to study the interaction of proteins involved in neurodegeneration.
Collapse
Affiliation(s)
| | | | | | - Sarah J. Annesley
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
18
|
Takaichi Y, Chambers JK, Ano Y, Takashima A, Nakayama H, Uchida K. Deposition of Phosphorylated α-Synuclein and Activation of GSK-3β and PP2A in the PS19 Mouse Model of Tauopathy. J Neuropathol Exp Neurol 2021; 80:731-740. [PMID: 34151989 DOI: 10.1093/jnen/nlab054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The simultaneous accumulation of multiple pathological proteins, such as hyperphosphorylated tau (hp-tau) and phosphorylated α-synuclein (p-αSyn), has been reported in the brains of patients with various neurodegenerative diseases. We previously demonstrated that hp-tau-dependent p-αSyn accumulation was associated with the activation of GSK-3β in the brains of P301L tau transgenic mice. To confirm the effects of another mutant tau on p-αSyn accumulation in vivo, we herein examined the brains of PS19 mice that overexpress human P301S mutant tau. Immunohistochemically, hp-tau and p-αSyn aggregates were detected in the same neuronal cells in the cerebrum and brain stem of aged PS19 mice. A semiquantitative analysis showed a positive correlation between hp-tau and p-αSyn accumulation. Furthermore, an activated form of GSK-3β was detected within cells containing both hp-tau and p-αSyn aggregates in PS19 mice. Western blotting showed a decrease in inactivated PP2A levels in PS19 mice. The present results suggest that the overexpression of human P301S mutant tau induces p-αSyn accumulation that is accompanied by not only GSK-3β, but also PP2A activation in PS19 mice, and highlight the synergic effects between tau and αSyn in the pathophysiology of neurodegenerative diseases that show the codeposition of tau and αSyn.
Collapse
Affiliation(s)
| | - James K Chambers
- From the Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo (YT, JKC, HN, KU); Research Laboratories for Health Science & Food Technologies and the Central Laboratories for Key Technologies, Kirin Company Ltd, Kanagawa (YA); Department of Life Science, Faculty of Science, Gakushuin University, Tokyo (AT), Japan
| | - Yasuhisa Ano
- From the Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo (YT, JKC, HN, KU); Research Laboratories for Health Science & Food Technologies and the Central Laboratories for Key Technologies, Kirin Company Ltd, Kanagawa (YA); Department of Life Science, Faculty of Science, Gakushuin University, Tokyo (AT), Japan
| | - Akihiko Takashima
- From the Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo (YT, JKC, HN, KU); Research Laboratories for Health Science & Food Technologies and the Central Laboratories for Key Technologies, Kirin Company Ltd, Kanagawa (YA); Department of Life Science, Faculty of Science, Gakushuin University, Tokyo (AT), Japan
| | - Hiroyuki Nakayama
- From the Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo (YT, JKC, HN, KU); Research Laboratories for Health Science & Food Technologies and the Central Laboratories for Key Technologies, Kirin Company Ltd, Kanagawa (YA); Department of Life Science, Faculty of Science, Gakushuin University, Tokyo (AT), Japan
| | - Kazuyuki Uchida
- From the Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo (YT, JKC, HN, KU); Research Laboratories for Health Science & Food Technologies and the Central Laboratories for Key Technologies, Kirin Company Ltd, Kanagawa (YA); Department of Life Science, Faculty of Science, Gakushuin University, Tokyo (AT), Japan
| |
Collapse
|
19
|
Interaction between Parkin and α-Synuclein in PARK2-Mediated Parkinson's Disease. Cells 2021; 10:cells10020283. [PMID: 33572534 PMCID: PMC7911026 DOI: 10.3390/cells10020283] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Parkin and α-synuclein are two key proteins involved in the pathophysiology of Parkinson's disease (PD). Neurotoxic alterations of α-synuclein that lead to the formation of toxic oligomers and fibrils contribute to PD through synaptic dysfunction, mitochondrial impairment, defective endoplasmic reticulum and Golgi function, and nuclear dysfunction. In half of the cases, the recessively inherited early-onset PD is caused by loss of function mutations in the PARK2 gene that encodes the E3-ubiquitin ligase, parkin. Parkin is involved in the clearance of misfolded and aggregated proteins by the ubiquitin-proteasome system and regulates mitophagy and mitochondrial biogenesis. PARK2-related PD is generally thought not to be associated with Lewy body formation although it is a neuropathological hallmark of PD. In this review article, we provide an overview of post-mortem neuropathological examinations of PARK2 patients and present the current knowledge of a functional interaction between parkin and α-synuclein in the regulation of protein aggregates including Lewy bodies. Furthermore, we describe prevailing hypotheses about the formation of intracellular micro-aggregates (synuclein inclusions) that might be more likely than Lewy bodies to occur in PARK2-related PD. This information may inform future studies aiming to unveil primary signaling processes involved in PD and related neurodegenerative disorders.
Collapse
|
20
|
Wiersma VI, Hoozemans JJM, Scheper W. Untangling the origin and function of granulovacuolar degeneration bodies in neurodegenerative proteinopathies. Acta Neuropathol Commun 2020; 8:153. [PMID: 32883341 PMCID: PMC7469111 DOI: 10.1186/s40478-020-00996-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
In the brains of tauopathy patients, tau pathology coincides with the presence of granulovacuolar degeneration bodies (GVBs) both at the regional and cellular level. Recently, it was shown that intracellular tau pathology causes GVB formation in experimental models thus explaining the strong correlation between these neuropathological hallmarks in the human brain. These novel models of GVB formation provide opportunities for future research into GVB biology, but also urge reevaluation of previous post-mortem observations. Here, we review neuropathological data on GVBs in tauopathies and other neurodegenerative proteinopathies. We discuss the possibility that intracellular aggregates composed of proteins other than tau are also able to induce GVB formation. Furthermore, the potential mechanisms of GVB formation and the downstream functional implications hereof are outlined in view of the current available data. In addition, we provide guidelines for the identification of GVBs in tissue and cell models that will help to facilitate and streamline research towards the elucidation of the role of these enigmatic and understudied structures in neurodegeneration.
Collapse
|
21
|
Tau and Alpha Synuclein Synergistic Effect in Neurodegenerative Diseases: When the Periphery Is the Core. Int J Mol Sci 2020; 21:ijms21145030. [PMID: 32708732 PMCID: PMC7404325 DOI: 10.3390/ijms21145030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023] Open
Abstract
In neuronal cells, tau is a microtubule-associated protein placed in axons and alpha synuclein is enriched at presynaptic terminals. They display a propensity to form pathologic aggregates, which are considered the underlying cause of Alzheimer's and Parkinson's diseases. Their functional impairment induces loss of axonal transport, synaptic and mitochondrial disarray, leading to a "dying back" pattern of degeneration, which starts at the periphery of cells. In addition, pathologic spreading of alpha-synuclein from the peripheral nervous system to the brain through anatomical connectivity has been demonstrated for Parkinson's disease. Thus, examination of the extent and types of tau and alpha-synuclein in peripheral tissues and their relation to brain neurodegenerative diseases is of relevance since it may provide insights into patterns of protein aggregation and neurodegeneration. Moreover, peripheral nervous tissues are easily accessible in-vivo and can play a relevant role in the early diagnosis of these conditions. Up-to-date investigations of tau species in peripheral tissues are scant and have mainly been restricted to rodents, whereas, more evidence is available on alpha synuclein in peripheral tissues. Here we aim to review the literature on the functional role of tau and alpha synuclein in physiological conditions and disease at the axonal level, their distribution in peripheral tissues, and discuss possible commonalities/diversities as well as their interaction in proteinopathies.
Collapse
|
22
|
Williams T, Sorrentino Z, Weinrich M, Giasson BI, Chakrabarty P. Differential cross-seeding properties of tau and α-synuclein in mouse models of tauopathy and synucleinopathy. Brain Commun 2020; 2:fcaa090. [PMID: 33094280 PMCID: PMC7567170 DOI: 10.1093/braincomms/fcaa090] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/01/2020] [Accepted: 06/08/2020] [Indexed: 11/17/2022] Open
Abstract
Co-occurrence of tau and α-synuclein pathologies in a subset of Alzheimer’s disease patients has led to the idea that mixed pathologies may play a unique characteristic role in the Alzheimer’s disease neurodegenerative cascade. To understand the aetiology of such mixed pathologies, we investigated cross-seeding by human recombinant tau and human recombinant α-synuclein fibrillar species in a mouse model of tauopathy (Line PS19) or synucleinopathy (Line M20). Unilateral hippocampal injection of tau fibrils or α-synuclein fibrils, and to a lesser extent tau + α-synuclein copolymer fibrils prepared from co-incubating individual recombinant monomers, induced robust phosphorylated tau pathology in PS19 mice relative to control mice. Though the tau + α-synuclein copolymer fibrils did not modulate induction of pathologies at the site of injection, examination of the whole brain showed that these copolymers exacerbated neuroanatomic transmission of seeded tau pathology compared to tau fibril-injected mice. Only α-synuclein fibrils, but not tau alone or tau + α-synuclein copolymers, triggered modest levels of endogenous phosphorylated α-synuclein pathology. Overall, data from the PS19 mice suggest that human α-synuclein fibrils can efficiently cross-seed human tau and have a modest priming effect on mouse α-synuclein, and the presence of tau fibrils does not exacerbate the priming process. In M20 mice, unilateral hippocampal injection of α-synuclein fibrils or tau fibrils induced robust bilateral phosphorylated α-synuclein pathology, while tau + α-synuclein copolymer injection resulted in restricted phosphorylated α-synuclein pathology predominantly in the ipsilateral cortex. This suggests that human tau fibrils can also induce human α-synuclein pathogenesis, and the presence of combinatorial seeds is not synergistic. None of these aggregates induced phosphorylated tau pathology in M20 mice, showing that mouse tau cannot be primed efficiently by human tau fibrils or human α-synuclein fibrils. Neuropathological analysis of the whole brain of M20 mice showed that tau + α-synuclein copolymer-injected mice had lower abundance of bilaterally transmitted α-synuclein pathologies relative to α-synuclein fibril-injected mice. Thus, the tau + α-synuclein copolymer fibrils show robust transmission properties preferentially in rodent model of tauopathies but not in synucleinopathy, probably signifying an enhanced cooperative relationship between tau and α-synuclein in the tau seeding process. Together, our data highlight the unique cross-seeding properties of tau and αSyn in neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- Tosha Williams
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Zachary Sorrentino
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Mary Weinrich
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
23
|
Brunello CA, Merezhko M, Uronen RL, Huttunen HJ. Mechanisms of secretion and spreading of pathological tau protein. Cell Mol Life Sci 2020; 77:1721-1744. [PMID: 31667556 PMCID: PMC7190606 DOI: 10.1007/s00018-019-03349-1] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
Accumulation of misfolded and aggregated forms of tau protein in the brain is a neuropathological hallmark of tauopathies, such as Alzheimer's disease and frontotemporal lobar degeneration. Tau aggregates have the ability to transfer from one cell to another and to induce templated misfolding and aggregation of healthy tau molecules in previously healthy cells, thereby propagating tau pathology across different brain areas in a prion-like manner. The molecular mechanisms involved in cell-to-cell transfer of tau aggregates are diverse, not mutually exclusive and only partially understood. Intracellular accumulation of misfolded tau induces several mechanisms that aim to reduce the cellular burden of aggregated proteins and also promote secretion of tau aggregates. However, tau may also be released from cells physiologically unrelated to protein aggregation. Tau secretion involves multiple vesicular and non-vesicle-mediated pathways, including secretion directly through the plasma membrane. Consequently, extracellular tau can be found in various forms, both as a free protein and in vesicles, such as exosomes and ectosomes. Once in the extracellular space, tau aggregates can be internalized by neighboring cells, both neurons and glial cells, via endocytic, pinocytic and phagocytic mechanisms. Importantly, accumulating evidence suggests that prion-like propagation of misfolding protein pathology could provide a general mechanism for disease progression in tauopathies and other related neurodegenerative diseases. Here, we review the recent literature on cellular mechanisms involved in cell-to-cell transfer of tau, with a particular focus in tau secretion.
Collapse
Affiliation(s)
- Cecilia A Brunello
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Riikka-Liisa Uronen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland.
| |
Collapse
|
24
|
Castillo-Carranza DL, Guerrero-Muñoz MJ, Sengupta U, Gerson JE, Kayed R. α-Synuclein Oligomers Induce a Unique Toxic Tau Strain. Biol Psychiatry 2018; 84:499-508. [PMID: 29478699 PMCID: PMC6201292 DOI: 10.1016/j.biopsych.2017.12.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND The coexistence of α-synuclein and tau aggregates in several neurodegenerative disorders, including Parkinson's disease and Alzheimer's disease, raises the possibility that a seeding mechanism is involved in disease progression. METHODS To further investigate the role of α-synuclein in the tau aggregation pathway, we performed a set of experiments using both recombinant and brain-derived tau and α-synuclein oligomers to seed monomeric tau aggregation in vitro and in vivo. Brain-derived tau oligomers were isolated from well-characterized cases of progressive supranuclear palsy (n = 4) and complexes of brain-derived α-synuclein/tau oligomers isolated from patients with Parkinson's disease (n = 4). The isolated structures were purified and characterized by standard biochemical methods, then injected into Htau mice (n = 24) to assess their toxicity and role in tau aggregation. RESULTS We found that α-synuclein induced a distinct toxic tau oligomeric strain that avoids fibril formation. In vivo, Parkinson's disease brain-derived α-synuclein/tau oligomers administered into Htau mouse brains accelerated endogenous tau oligomer formation concurrent with increasing cell loss. CONCLUSIONS Our findings provide evidence, for the first time, that α-synuclein enhances the harmful effects of tau, thus contributing to disease progression.
Collapse
Affiliation(s)
| | | | | | | | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
25
|
Daniele S, Frosini D, Pietrobono D, Petrozzi L, Lo Gerfo A, Baldacci F, Fusi J, Giacomelli C, Siciliano G, Trincavelli ML, Franzoni F, Ceravolo R, Martini C, Bonuccelli U. α-Synuclein Heterocomplexes with β-Amyloid Are Increased in Red Blood Cells of Parkinson's Disease Patients and Correlate with Disease Severity. Front Mol Neurosci 2018; 11:53. [PMID: 29520218 PMCID: PMC5827358 DOI: 10.3389/fnmol.2018.00053] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/07/2018] [Indexed: 02/02/2023] Open
Abstract
Neurodegenerative disorders (NDs) are characterized by abnormal accumulation/misfolding of specific proteins, primarily α-synuclein (α-syn), β-amyloid1-42 (Aβ1-42) and tau, in both brain and peripheral tissues. In addition to oligomers, the role of the interactions of α-syn with Aβ or tau has gradually emerged. Nevertheless, despite intensive research, NDs have no accepted peripheral markers for biochemical diagnosis. In this respect, Red Blood Cells (RBCs) are emerging as a valid peripheral model for the study of aging-related pathologies. Herein, a small cohort (N = 28) of patients affected by Parkinson's disease (PD) and age-matched controls were enrolled to detect the content of α-syn (total and oligomeric), Aβ1-42 and tau (total and phosphorylated) in RBCs. Moreover, the presence of α-syn association with tau and Aβ1-42 was explored by co-immunoprecipitation/western blotting in the same cells, and quantitatively confirmed by immunoenzymatic assays. For the first time, PD patients were demonstrated to exhibit α-syn heterocomplexes with Aβ1-42 and tau in peripheral tissues; interestingly, α-syn-Aβ1-42 concentrations were increased in PD subjects with respect to healthy controls (HC), and directly correlated with disease severity and motor deficits. Moreover, total-α-syn levels were decreased in PD subjects and inversely related to their motor deficits. Finally, an increase of oligomeric-α-syn and phosphorylated-tau was observed in RBCs of the enrolled patients. The combination of three parameters (total-α-syn, phosphorylated-tau and α-syn-Aβ1-42 concentrations) provided the best fitting predictive index for discriminating PD patients from controls. Nevertheless further investigations should be required, overall, these data suggest α-syn hetero-aggregates in RBCs as a putative tool for the diagnosis of PD.
Collapse
Affiliation(s)
| | - Daniela Frosini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Lucia Petrozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Annalisa Lo Gerfo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
26
|
Spires-Jones TL, Attems J, Thal DR. Interactions of pathological proteins in neurodegenerative diseases. Acta Neuropathol 2017; 134:187-205. [PMID: 28401333 PMCID: PMC5508034 DOI: 10.1007/s00401-017-1709-7] [Citation(s) in RCA: 296] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 02/06/2023]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), frontotemporal lobar degeneration (FTD), Lewy body disease (LBD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) have in common that protein aggregates represent pathological hallmark lesions. Amyloid β-protein, τ-protein, α-synuclein, and TDP-43 are the most frequently aggregated proteins in these disorders. Although they are assumed to form disease-characteristic aggregates, such as amyloid plaques and neurofibrillary tangles in AD or Lewy bodies in LBD/PD, they are not restricted to these clinical presentations. They also occur in non-diseased individuals and can co-exist in the same brain without or with a clinical picture of a distinct dementing or movement disorder. In this review, we discuss the co-existence of these pathologies and potential additive effects in the human brain as well as related functional findings on cross-seeding and molecular interactions between these aggregates/proteins. We conclude that there is evidence for interactions at the molecular level as well as for additive effects on brain damage by multiple pathologies occurring in different functionally important neurons. Based upon this information, we hypothesize a cascade of events that may explain general mechanisms in the development of neurodegenerative disorders: (1) distinct lesions are a prerequisite for the development of a distinct disease (e.g., primary age-related tauopathy for AD), (2) disease-specific pathogenic events further trigger the development of a specific disease (e.g., Aβ aggregation in AD that exaggerate further Aβ and AD-related τ pathology), (3) the symptomatic disease manifests, and (4) neurodegenerative co-pathologies may be either purely coincidental or (more likely) have influence on the disease development and/or its clinical presentation.
Collapse
Affiliation(s)
- Tara L Spires-Jones
- Centre for Dementia Prevention, and Euan MacDonald Centre for Motor Neurone Disease, The University of Edinburgh Centre for Cognitive and Neural Systems, 1 George Square, Edinburgh, EH8 9JZ, UK.
| | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Dietmar Rudolf Thal
- Departement Neurowetenschappen, Katholieke Universiteit Leuven, Herestraat 49, 3000, Leuven, Belgium
- Departement Pathologische Ontleedkunde, UZ Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
27
|
Perez-Soriano A, Arena JE, Dinelle K, Miao Q, McKenzie J, Neilson N, Puschmann A, Schaffer P, Shinotoh H, Smith-Forrester J, Shahinfard E, Vafai N, Wile D, Wszolek Z, Higuchi M, Sossi V, Stoessl AJ. PBB3 imaging in Parkinsonian disorders: Evidence for binding to tau and other proteins. Mov Disord 2017; 32:1016-1024. [DOI: 10.1002/mds.27029] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/25/2017] [Accepted: 03/30/2017] [Indexed: 01/06/2023] Open
Affiliation(s)
- Alexandra Perez-Soriano
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
| | - Julieta E. Arena
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
| | - Katie Dinelle
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
- Department of Physics & Astronomy; University of British Columbia; Vancouver BC Canada
| | | | - Jessamyn McKenzie
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
| | - Nicole Neilson
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
| | - Andreas Puschmann
- Department of Clinical Sciences; Lund University, Skåne University Hospital; Lund Sweden
| | | | | | - Jenna Smith-Forrester
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
- Department of Physics & Astronomy; University of British Columbia; Vancouver BC Canada
| | - Elham Shahinfard
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
- Department of Physics & Astronomy; University of British Columbia; Vancouver BC Canada
| | - Nasim Vafai
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
- Department of Physics & Astronomy; University of British Columbia; Vancouver BC Canada
| | - Daryl Wile
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
| | | | - Makoto Higuchi
- National Institute of Radiological Sciences; Chiba Japan
| | - Vesna Sossi
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
- Department of Physics & Astronomy; University of British Columbia; Vancouver BC Canada
| | - A. Jon Stoessl
- Pacific Parkinson's Research Centre; Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia & Vancouver Coastal Health; Vancouver BC Canada
| |
Collapse
|
28
|
Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol 2017; 133:665-704. [PMID: 28386764 PMCID: PMC5390006 DOI: 10.1007/s00401-017-1707-9] [Citation(s) in RCA: 642] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/26/2017] [Accepted: 03/26/2017] [Indexed: 01/18/2023]
Abstract
Tau is well established as a microtubule-associated protein in neurons. However, under pathological conditions, aberrant assembly of tau into insoluble aggregates is accompanied by synaptic dysfunction and neural cell death in a range of neurodegenerative disorders, collectively referred to as tauopathies. Recent advances in our understanding of the multiple functions and different locations of tau inside and outside neurons have revealed novel insights into its importance in a diverse range of molecular pathways including cell signalling, synaptic plasticity, and regulation of genomic stability. The present review describes the physiological and pathophysiological properties of tau and how these relate to its distribution and functions in neurons. We highlight the post-translational modifications of tau, which are pivotal in defining and modulating tau localisation and its roles in health and disease. We include discussion of other pathologically relevant changes in tau, including mutation and aggregation, and how these aspects impinge on the propensity of tau to propagate, and potentially drive neuronal loss, in diseased brain. Finally, we describe the cascade of pathological events that may be driven by tau dysfunction, including impaired axonal transport, alterations in synapse and mitochondrial function, activation of the unfolded protein response and defective protein degradation. It is important to fully understand the range of neuronal functions attributed to tau, since this will provide vital information on its involvement in the development and pathogenesis of disease. Such knowledge will enable determination of which critical molecular pathways should be targeted by potential therapeutic agents developed for the treatment of tauopathies.
Collapse
Affiliation(s)
- Tong Guo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK
| | - Diane P Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK.
| |
Collapse
|
29
|
The Role of α-Synuclein and LRRK2 in Tau Phosphorylation. PARKINSONS DISEASE 2015; 2015:734746. [PMID: 25977830 PMCID: PMC4419261 DOI: 10.1155/2015/734746] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 01/08/2023]
Abstract
There is now a considerable body of experimental evidence that Parkinson's disease arises through physiological interaction of causative molecules, leading to tau pathology. In this review, we discuss the physiological role of α-synuclein and LRRK2 in the abnormal phosphorylation of tau. In addition, as recent reports have indicated that heat shock proteins- (HSPs-) inducing drugs can help to ameliorate neurodegenerative diseases associated with tau pathology, we also discuss therapeutic strategies for PD focusing on inhibition of α-synuclein- and LRRK2-associated tau phosphorylation by HSPs.
Collapse
|
30
|
Alpha-synuclein and tau: teammates in neurodegeneration? Mol Neurodegener 2014; 9:43. [PMID: 25352339 PMCID: PMC4230508 DOI: 10.1186/1750-1326-9-43] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 10/16/2014] [Indexed: 11/25/2022] Open
Abstract
The accumulation of α-synuclein aggregates is the hallmark of Parkinson’s disease, and more generally of synucleinopathies. The accumulation of tau aggregates however is classically found in the brains of patients with dementia, and this type of neuropathological feature specifically defines the tauopathies. Nevertheless, in numerous cases α-synuclein positive inclusions are also described in tauopathies and vice versa, suggesting a co-existence or crosstalk of these proteinopathies. Interestingly, α-synuclein and tau share striking common characteristics suggesting that they may work in concord. Tau and α-synuclein are both partially unfolded proteins that can form toxic oligomers and abnormal intracellular aggregates under pathological conditions. Furthermore, mutations in either are responsible for severe dominant familial neurodegeneration. Moreover, tau and α-synuclein appear to promote the fibrillization and solubility of each other in vitro and in vivo. This suggests that interactions between tau and α-synuclein form a deleterious feed-forward loop essential for the development and spreading of neurodegeneration. Here, we review the recent literature with respect to elucidating the possible links between α-synuclein and tau.
Collapse
|
31
|
Confluence of α-synuclein, tau, and β-amyloid pathologies in dementia with Lewy bodies. J Neuropathol Exp Neurol 2014; 72:1203-12. [PMID: 24226269 DOI: 10.1097/nen.0000000000000018] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dementia with Lewy bodies (DLB) is pathologically characterized by α-synuclein aggregates in the brain. Most patients with DLB also show cerebral Alzheimer disease-type pathology (i.e. β-amyloid plaques and hyperphosphorylated tau deposits). It is unclear whether this overlap is coincidental or driven by specific regional or cellular interactions. The aims of this study were to investigate the regional convergence of α-synuclein, tau, and β-amyloid and to identify patterns of cellular co-occurrence of tau and α-synuclein in DLB. The study group consisted of 22 patients who met clinical and neuropathologic criteria for DLB. Protein aggregates were assessed semiquantitatively in 17 brain areas. APOE and MAPT genotypes were determined. Cellular co-occurrence of tau and α-synuclein was evaluated by double immunofluorescence. We found that total β-amyloid pathology scores correlated positively with total α-synuclein pathology scores (ρ = 0.692, p = 0.001). The factors that correlated best with the amount of α-synuclein pathology were the severity of β-amyloid pathology and presence of the MAPT H1 haplotype. Tau and α-synuclein frequently colocalized in limbic areas, but no correlation between total pathology scores was observed. This study confirms and extends the role of β-amyloid deposition and the MAPT H1 haplotype as contributing factors in DLB pathogenesis and demonstrates the confluence of multiple agents in neurodegenerative diseases.
Collapse
|
32
|
Nagaishi M, Yokoo H, Nakazato Y. Tau-positive glial cytoplasmic granules in multiple system atrophy. Neuropathology 2010; 31:299-305. [PMID: 21062361 DOI: 10.1111/j.1440-1789.2010.01159.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multiple system atrophy (MSA) is a sporadic neurodegenerative disease that is pathologically characterized by the filamentous aggregation of α-synuclein. We report a case of MSA showing unusual neuropathological findings and review six autopsied cases of MSA. The patient progressively developed parkinsonism and ataxia for the 9 years prior to her death at the age of 72 years. Neuropathological examinations revealed neuronal loss restricted to the olivopontocerebellar and striatonigral region, which was more severe in the putamen. Staining with anti-α-synuclein antibody demonstrated widespread occurrence of glial cytoplasmic inclusions, which mainly accumulated in oligodendroglial cells and corresponded closely to the degree of disease progression. In addition, tau-positive granules were detected within the glial cytoplasm in the neurodegenerative region, which was especially prominent in the putamen and internal capsule. Tau accumulation was also clearly recognized by staining with specific antibodies against three-repeat or four-repeat tau. The glia that demonstrated deposition of tau-positive granules were distinguished from α-synuclein-positive oligodendroglia by double immunohistochemical staining. These characteristic glial accumulations of tau were also present in all six cases of MSA. These results indicate that tau-positive granules in glia are common findings in MSA and that tau aggregation might be another pathway to neurodegeneration in MSA.
Collapse
Affiliation(s)
- Masaya Nagaishi
- Department of Human Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.
| | | | | |
Collapse
|
33
|
Early-onset familial lewy body dementia with extensive tauopathy: a clinical, genetic, and neuropathological study. J Neuropathol Exp Neurol 2009; 68:73-82. [PMID: 19104444 DOI: 10.1097/nen.0b013e3181927577] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We describe a Spanish family in which 3 of 4 siblings had dementia with Lewy bodies, 2 of them starting at age 26 years and the other at 29 years. The father has recently been diagnosed with Lewy body disease, with onset at 77 years. Neuropathological examination of the brain of the index patient disclosed unusual features characterized by diffuse Lewy body disease and generalized neurofibrillary tangle pathology but with no amyloid deposits in any region. Moreover, Lewy body pathology colocalized with neurofibrillary tangles in most affected neurons. Mutation screening that included all coding exons of presenilin 1 (PSEN1), presenilin 2 (PSEN2), alpha-synuclein (SNCA), beta-synuclein (SNCB), microtubule-associated protein tau (MAPT), leucine-rich repeat kinase 2 (LRRK2), glucocerebrosidase (GBA), and exons 16 and 17 of the amyloid precursor protein (APP) genes did not identify any mutation. Genome-wide single nucleotide polymorphism was performed in 4 family members and ruled out any pathogenic duplication or deletion in the entire genome. In summary, we report a unique family with pathologically confirmed early-onset dementia with Lewy bodies with widespread tau and alpha-synuclein deposition. The absence of mutations in genes known to cause Lewy body disease suggests that a novel locus or loci are implicated in this neurodegenerative disease.
Collapse
|
34
|
Hasegawa M, Arai T, Nonaka T, Kametani F, Yoshida M, Hashizume Y, Beach TG, Buratti E, Baralle F, Morita M, Nakano I, Oda T, Tsuchiya K, Akiyama H. Phosphorylated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Ann Neurol 2008; 64:60-70. [PMID: 18546284 PMCID: PMC2674108 DOI: 10.1002/ana.21425] [Citation(s) in RCA: 606] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE TAR DNA-binding protein of 43kDa (TDP-43) is deposited as cytoplasmic and intranuclear inclusions in brains of patients with frontotemporal lobar degeneration with ubiquitinated inclusions (FTLD-U) and amyotrophic lateral sclerosis (ALS). Previous studies reported that abnormal phosphorylation takes place in deposited TDP-43. The aim of this study was to identify the phosphorylation sites and responsible kinases, and to clarify the pathological significance of phosphorylation of TDP-43. METHODS We generated multiple antibodies specific to phosphorylated TDP-43 by immunizing phosphopeptides of TDP-43, and analyzed FTLD-U and ALS brains by immunohistochemistry, immunoelectron microscopy, and immunoblots. In addition, we performed investigations aimed at identifying the responsible kinases, and we assessed the effects of phosphorylation on TDP-43 oligomerization and fibrillization. RESULTS We identified multiple phosphorylation sites in carboxyl-terminal regions of deposited TDP-43. Phosphorylation-specific antibodies stained more inclusions than antibodies to ubiquitin and, unlike existing commercially available anti-TDP-43 antibodies, did not stain normal nuclei. Ultrastructurally, these antibodies labeled abnormal fibers of 15nm diameter and on immunoblots recognized hyperphosphorylated TDP-43 at 45kDa, with additional 18 to 26kDa fragments in sarkosyl-insoluble fractions from FTLD-U and ALS brains. The phosphorylated epitopes were generated by casein kinase-1 and -2, and phosphorylation led to increased oligomerization and fibrillization of TDP-43. INTERPRETATION These results suggest that phosphorylated TDP-43 is a major component of the inclusions, and that abnormal phosphorylation of TDP-43 is a critical step in the pathogenesis of FTLD-U and ALS. Phosphorylation-specific antibodies will be powerful tools for the investigation of these disorders.
Collapse
Affiliation(s)
- Masato Hasegawa
- Department of Molecular Neurobiology, Tokyo Institute of Psychiatry, Tokyo Metropolitan Organization for Medical Research, Kamikitazawa, Setagaya-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Fujishiro H, Tsuboi Y, Lin WL, Uchikado H, Dickson DW. Co-localization of tau and alpha-synuclein in the olfactory bulb in Alzheimer's disease with amygdala Lewy bodies. Acta Neuropathol 2008; 116:17-24. [PMID: 18446351 PMCID: PMC2719249 DOI: 10.1007/s00401-008-0383-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Revised: 04/19/2008] [Accepted: 04/20/2008] [Indexed: 11/24/2022]
Abstract
We recently reported that Alzheimer's disease (AD) with amygdala Lewy bodies (ALB) is a distinct form of alpha-synucleinopathy that occurs in advanced AD. In AD/ALB the alpha-synuclein pathology correlated with tau pathology, but not amyloid plaques, and there was often co-localization of tau and alpha-synuclein in the same neuron. Given the anatomical connectivity of the anterior olfactory nucleus and the amygdala, which receives axonal projections from the olfactory bulb, we hypothesized that there might be a relationship between tau and alpha-synuclein pathology in the olfactory bulb and the amygdala in AD. We screened for alpha-synuclein pathology in the olfactory bulb in AD with and without ALB, and investigated its relationship with tau pathology. In 38 of 41 (93%) AD/ALB cases and 4 of 21 (19%) AD cases without ALB (AD/non-ALB), alpha-synuclein pathology was detected in the olfactory bulb. Double immunolabeling at the light and electron microscopic levels revealed co-localization of tau and alpha-synuclein in the olfactory bulb neurons and neurites. The severity of tau pathology correlated with alpha-synuclein pathology in the olfactory bulb. In addition, alpha-synuclein pathology in the olfactory bulb correlated with alpha-synuclein pathology in amygdala. Tau pathology was greater in both the olfactory bulb and amygdala in AD/ALB than in AD/non-ALB, but there was no difference in tau pathology between the two groups in other brain regions assessed. The present study shows that in AD/ALB, the olfactory bulb is nearly equally vulnerable to tau and alpha-synuclein pathology as the amygdala and suggests that neurodegeneration in these two anatomical regions is linked.
Collapse
Affiliation(s)
- Hiroshige Fujishiro
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | | | | | | |
Collapse
|
36
|
Bowen S, Ateh DD, Deinhardt K, Bird MM, Price KM, Baker CS, Robson JC, Swash M, Shamsuddin W, Kawar S, El-Tawil T, Roos J, Hoyle A, Nickols CD, Knowles CH, Pullen AH, Luthert PJ, Weller RO, Hafezparast M, Franklin RJM, Revesz T, King RHM, Berninghausen O, Fisher EMC, Schiavo G, Martin JE. The phagocytic capacity of neurones. Eur J Neurosci 2007; 25:2947-55. [PMID: 17561810 DOI: 10.1111/j.1460-9568.2007.05554.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Phagocytosis is defined as the ingestion of particulates over 0.5 microm in diameter and is associated with cells of the immune system such as macrophages or monocytes. Neurones are not generally recognized to be phagocytic. Using light, confocal, time-lapse and electron microscopy, we carried out a wide range of in-vitro and in-vivo experiments to examine the phagocytic capacity of different neuronal cell types. We demonstrated phagocytosis of material by neurones, including cell debris and synthetic particles up to 2.8 microm in diameter. We showed phagocytosis in different neuronal types, and demonstrated that debris can be transported from neurite extremities to cell bodies and persist within neurones. Flow cytometry analysis demonstrated the lack of certain complement receptors on neurones but the presence of others, including integrin receptors known to mediate macrophage phagocytosis, indicating that a restricted set of phagocytosis receptors may mediate this process. Neuronal phagocytosis occurs in vitro and in vivo, and we propose that this is a more widespread and significant process than previously recognized. Neuronal phagocytosis may explain certain inclusions in neurones during disease, cell-to-cell spread of disease, neuronal death during disease progression and provide a potential mechanism for therapeutic intervention through the delivery of particulate drug carriers.
Collapse
Affiliation(s)
- Samantha Bowen
- Neuroscience Centre and Pathology Group, Queen Mary's School of Medicine and Dentistry, Institute of Pathology, Royal London Hospital, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yu S, Li X, Liu G, Han J, Zhang C, Li Y, Xu S, Liu C, Gao Y, Yang H, Uéda K, Chan P. Extensive nuclear localization of alpha-synuclein in normal rat brain neurons revealed by a novel monoclonal antibody. Neuroscience 2007; 145:539-55. [PMID: 17275196 DOI: 10.1016/j.neuroscience.2006.12.028] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 11/14/2006] [Accepted: 12/08/2006] [Indexed: 11/22/2022]
Abstract
Synuclein was initially named for its localization in both presynaptic nerve terminals and portions of nuclear envelope. However, subsequent studies only confirmed the presynaptic localization of this protein in the brain; its nuclear localization in the neurons remained elusive. Here, two new monoclonal antibodies against alpha-synuclein (alpha-SYN) were produced. Epitope mapping using phage peptide display showed that the epitopes of the two antibodies were localized in two distinct specific sequences of the C-terminal domain of alpha-SYN. One antibody named 3D5 recognized amino acids 115-121 of alpha-SYN and the other antibody named 2E3 identified the amino acids 134-138 of the protein. Western blot analysis demonstrated that both 2E3 and 3D5 detected a 19 kD protein from rat and human brain homogenates, which was identical to the molecular size of recombinant alpha-SYN. However, immunohistochemical staining on normal adult rat brain sections showed that the two antibodies revealed distinct patterns of subcellular localization of alpha-SYN immunoreactivity. Both 3D5 and 2E3 detected the presynaptic alpha-SYN but only 3D5 detected the nuclear alpha-SYN. The nuclear localization of alpha-SYN was further confirmed by Western blot analysis in isolated nuclear fraction where the same size of alpha-SYN was detected, and by immunoelectron microscopy using colloidal gold probes where gold particles were specifically localized in portions of peri- and intra-nucleus. The nuclear positive neurons were distributed extensively in almost all the brain regions. This is the first report well characterizing the extensive localization of alpha-SYN in the neuronal nuclei throughout the brain in normal conditions. This finding indicates an important physiological function of this molecule in the nuclei of brain neurons, which deserves further investigations.
Collapse
Affiliation(s)
- S Yu
- Key Laboratory on Neurodegenerative Diseases of Ministry of Education, Beijing Institute of Geriatrics and Xuanwu Hospital of the Capital University of Medical Sciences, Beijing 100053, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
During over 50 years of the first author's career in neuropathology at Montefiore Medical Center in New York, we have come across certain interesting neuropathological findings. In this communication, some photographs showing macroscopic, microscopic and electron microscopic significant findings are selected to illustrate usefulness not only for the diagnosis but also for understanding of the nervous system. The six topics presented in this paper are: (i) unattached presynaptic terminals in cerebellar neuroblastoma; (ii) neurofibrillary tangle formation in the nucleus basalis of Meynert ipsilateral to a massive cerebral infarct; (iii) orderly arrangement of tumor cells in leptomeningeal carcinomatosis; (iv) interface between craniopharyngioma and brain tissue; (v) neurofibrillary tangles and Lewy bodies in a single neuron; and (vi) Cu/Zn superoxide dismutase positive Lewy body-like hyaline inclusions in anterior horn cells in familial motor neuron diseases. Analyses of these findings are presented for an educational purpose.
Collapse
Affiliation(s)
- Asao Hirano
- Division of Neuropathology, Montefiore Medical Center, Bronx, New York, NY 10467-2490, USA.
| | | |
Collapse
|
39
|
Ardley HC, Robinson PA. The role of ubiquitin-protein ligases in neurodegenerative disease. NEURODEGENER DIS 2006; 1:71-87. [PMID: 16908979 DOI: 10.1159/000080048] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2003] [Accepted: 02/19/2004] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease and Parkinson's disease are the most common neurodegenerative conditions associated with the ageing process. The pathology of these and other neurodegenerative disorders, including polyglutamine diseases, is characterised by the presence of inclusion bodies in brain tissue of affected patients. In general, these inclusion bodies consist of insoluble, unfolded proteins that are commonly tagged with the small protein, ubiquitin. Covalent tagging of proteins with chains of ubiquitin generally targets them for degradation. Indeed, the ubiquitin/proteasome system (UPS) is the major route through which intracellular proteolysis is regulated. This strongly implicates the UPS in these disease-associated inclusions, either due to malfunction (of specific UPS components) or overload of the system (due to aggregation of unfolded/mutant proteins), resulting in subsequent cellular toxicity. Protein targeting for degradation is a highly regulated process. It relies on transfer of ubiquitin molecules to the target protein via an enzyme cascade and specific recognition of a substrate protein by ubiquitin-protein ligases (E3s). Recent advances in our knowledge gained from the Human Genome Mapping Project have revealed the presence of potentially hundreds of E3s within the human genome. The discovery that parkin, mutations in which are found in at least 50% of patients with autosomal recessive juvenile parkinsonism, is an E3 further highlights the importance of the UPS in neurological disease. To date, parkin is the only E3 confirmed to have a direct causal role in neurodegenerative disorders. However, a number of other (putative) E3s have now been identified that may cause disease directly or interact with neurological disease-associated proteins. Many of these are either lost or mutated in a given disease or fail to process disease-associated mutant proteins correctly. In this review, we will discuss the role(s) of E3s in neurodegenerative disorders.
Collapse
Affiliation(s)
- Helen C Ardley
- Molecular Medicine Unit, University of Leeds, St. James's University Hospital, Leeds, UK.
| | | |
Collapse
|
40
|
Mukaetova-Ladinska EB, McKeith IG. Pathophysiology of synuclein aggregation in Lewy body disease. Mech Ageing Dev 2006; 127:188-202. [PMID: 16297436 DOI: 10.1016/j.mad.2005.09.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2004] [Revised: 03/15/2005] [Accepted: 09/15/2005] [Indexed: 10/25/2022]
Abstract
We provide an overview of synaptic pathology in dementia with Lewy bodies (DLB) and related neurodegenerative disorders that are characterised by intraneuronal accumulation of alpha-synuclein aggregates. The review addresses the clinico-neuropathological correlates of synaptic pathology in Lewy body disease, and concentrates on: altered alpha-synuclein metabolism, mechanisms leading to alpha-synuclein fibril formation (self-polymerisation, alpha-synuclein mutations and post-translational modifications) and how these influence the axonal transport and synaptic network in ageing and disease process. Understanding the mechanisms leading to intraneuronal alpha-synuclein accumulation are crucial for the development of novel therapies for treatment of Lewy body disease.
Collapse
Affiliation(s)
- Elizabeta B Mukaetova-Ladinska
- Institute for Ageing and Health, University of Newcastle, Newcastle General Hospital, Westgate Road, Newcastle upon Tyne NE4 6BE, UK.
| | | |
Collapse
|
41
|
Wenning GK, Jellinger KA. The role of α-synuclein and tau in neurodegenerative movement disorders. Curr Opin Neurol 2005; 18:357-62. [PMID: 16003109 DOI: 10.1097/01.wco.0000168241.53853.32] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Gregor K Wenning
- Department of Neurology, Medical University, Innsbruck, Austria.
| | | |
Collapse
|
42
|
Broe M, Shepherd CE, Mann DMA, Milward EA, Gai WP, Thiel E, Halliday GM. Insoluble alpha-synuclein in Alzheimer's disease without Lewy body formation. Neurotox Res 2005; 7:69-76. [PMID: 15639799 DOI: 10.1007/bf03033777] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Insoluble alpha-synuclein plays a central role in Lewy body diseases, with considerable controversy as to whether it plays a similar role in Alzheimer's disease (AD). We assessed the tissue location and solubility of cortical alpha-synuclein in AD (without Lewy body formation) compared with controls, using sequential extraction procedures and Western immunoblotting to quantify different alpha-synuclein species in their different solubility states. Controls had no insoluble cortical alpha-synuclein and a ratio of soluble:lipid-associated alpha-synuclein of 1.2-/+0.1. Total alpha-synuclein protein was significantly increased in AD and concentrated within the lipid-associated fraction (soluble:lipid ratio 0.9-/+0.05, soluble:insoluble 1.5-/+0.1, lipid:insoluble 1.7-/+0.1) which proved difficult to localize in paraffin-embedded tissue. Tissues prepared without lipid extraction revealed alpha-synuclein-immunoreactivity in the amorphous components of mature cored AD plaques. This lipid-association of alpha-synuclein in mature AD plaques links this protein with other lipid changes thought to be important in disease pathogenesis.
Collapse
Affiliation(s)
- Melissa Broe
- Prince of Wales Medical Research Institute, Sydney, 2031 Australia
| | | | | | | | | | | | | |
Collapse
|
43
|
Totterdell S, Hanger D, Meredith GE. The ultrastructural distribution of alpha-synuclein-like protein in normal mouse brain. Brain Res 2004; 1004:61-72. [PMID: 15033420 DOI: 10.1016/j.brainres.2003.10.072] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2003] [Indexed: 10/26/2022]
Abstract
The synaptic protein alpha-synuclein is found throughout the brain, although its function remains ill-defined. Abnormal accumulations of alpha-synuclein have been recognised to be associated with a number of neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Nevertheless, little is known about the precise localisation of this protein within the normal brain, information which might contribute to our understanding of its role in both health and disease. We raised an antibody which recognises both human and murine alpha-synuclein and this was used to study the distribution of the protein in the normal mouse brain. We used morphological characteristics to classify the immunopositive presynaptic elements and their targets. We conclude that the protein is present in synaptic boutons of axons with different neurochemical phenotypes but that it is not present in all synaptic terminals. Furthermore, the protein is present in the terminals of neurons such as the dopaminergic neurons of the substantia nigra and the glutamatergic neurons of the hippocampus, cell types which accumulate alpha-synuclein in disease. Nevertheless alpha-synuclein is also found in terminals of neurons which have not been reported to accumulate the protein in neurodegenerative disorders.
Collapse
Affiliation(s)
- Susan Totterdell
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | | | | |
Collapse
|
44
|
Trimmer PA, Borland MK, Keeney PM, Bennett JP, Parker WD. Parkinson's disease transgenic mitochondrial cybrids generate Lewy inclusion bodies. J Neurochem 2004; 88:800-12. [PMID: 14756800 DOI: 10.1046/j.1471-4159.2003.02168.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Many models of Parkinson's disease (PD) have succeeded in replicating dopaminergic neuron loss or alpha-synuclein aggregation but not the formation of classical Lewy bodies, the pathological hallmark of PD. Our cybrid model of sporadic PD was created by introducing the mitochondrial genes from PD patients into neuroblastoma cells that lack mitochondrial DNA. Previous studies using cybrids have shown that information encoded by mitochondrial DNA in patients contributes to many pathogenic features of sporadic PD. In this paper, we report the generation of fibrillar and vesicular inclusions in a long-term cybrid cell culture model that replicates the essential antigenic and structural features of Lewy bodies in PD brain without the need for exogenous protein expression or inhibition of mitochondrial or proteasomal function. The inclusions generated by PD cybrid cells stained with eosin, thioflavin S, and antibodies to alpha-synuclein, ubiquitin, parkin, synphilin-1, neurofilament, beta-tubulin, the proteasome, nitrotyrosine, and cytochrome c. Future studies of these cybrids will enable us to better understand how Lewy bodies form and what role they play in the pathogenesis of PD.
Collapse
Affiliation(s)
- Patricia A Trimmer
- Center for the Study of Neurodegenerative Diseases and Department of Neurology, University of Virginia, School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | |
Collapse
|
45
|
Rademakers R, Cruts M, van Broeckhoven C. The role of tau (MAPT) in frontotemporal dementia and related tauopathies. Hum Mutat 2004; 24:277-95. [PMID: 15365985 DOI: 10.1002/humu.20086] [Citation(s) in RCA: 239] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Tau is a multifunctional protein that was originally identified as a microtubule-associated protein. In patients diagnosed with frontotemporal dementia and parkinsonism linked to chromosome 17, mutations in the gene encoding tau (MAPT) have been identified that disrupt the normal binding of tau to tubulin resulting in pathological deposits of hyperphosphorylated tau. Abnormal filamentous tau deposits have been reported as a pathological characteristic in several other neurodegenerative diseases, including frontotemporal dementia, Pick Disease, Alzheimer disease, argyrophilic grain disease, progressive supranuclear palsy, and corticobasal degeneration. In the last five years, extensive research has identified 34 different pathogenic MAPT mutations in 101 families worldwide. In vitro, cell-free and transfected cell studies have provided valuable information on tau dysfunction and transgenic mice carrying human MAPT mutations are being generated to study the influence of MAPT mutations in vivo. This mutation update describes the considerable differences in clinical and pathological presentation of patients with MAPT mutations and summarizes the effect of the different mutations on tau functioning. In addition, the role of tau as a genetic susceptibility factor is discussed, together with the genetic evidence for additional causal genes for tau-positive as well as tau-negative dementia.
Collapse
Affiliation(s)
- R Rademakers
- Department of Molecular Genetics, Neurogenetics Group, Flanders Interuniversity Institute for Biotechnology, University of Antwerp, Antwerpen, Belgium
| | | | | |
Collapse
|
46
|
Ardley HC, Scott GB, Rose SA, Tan NGS, Markham AF, Robinson PA. Inhibition of proteasomal activity causes inclusion formation in neuronal and non-neuronal cells overexpressing Parkin. Mol Biol Cell 2003; 14:4541-56. [PMID: 12937272 PMCID: PMC266771 DOI: 10.1091/mbc.e03-02-0078] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Association between protein inclusions and neurodegenerative diseases, including Parkinson's and Alzheimer's diseases, and polyglutamine disorders, has been widely documented. Although ubiquitin is conjugated to many of these aggregated proteins, the 26S proteasome does not efficiently degrade them. Mutations in the ubiquitin-protein ligase Parkin are associated with autosomal recessive juvenile Parkinsonism. Although Parkin-positive inclusions are not detected in brains of autosomal recessive juvenile Parkinsonism patients, Parkin is found in Lewy bodies in sporadic disease. This suggests that loss of Parkin ligase activity via mutation, or sequestration to Lewy bodies, is a contributory factor to sporadic disease onset. We now demonstrate that decreased proteasomal activity causes formation of large, noncytotoxic inclusions within the cytoplasm of both neuronal and nonneuronal cells overexpressing Parkin. This is not a general phenomenon as there is an absence of similar inclusions when HHARI, a structural homolog of Parkin, is overexpressed. The inclusions colocalize with ubiquitin and with proteasomes. Furthermore, Parkin inclusions colocalize with gamma-tubulin, acetylated alpha-tubulin, and cause redistribution of vimentin, suggesting aggresome-like properties. Our data imply that lower proteasomal activity, previously observed in brain tissue of Parkinson's disease patients, leads to Parkin accumulation and a concomitant reduction in ligase activity, thereby promoting Lewy body formation.
Collapse
Affiliation(s)
- Helen C Ardley
- Molecular Medicine Unit, University of Leeds, St. James's University Hospital, Leeds LS9 7TF, United Kingdom.
| | | | | | | | | | | |
Collapse
|
47
|
Hishikawa N, Hashizume Y, Ujihira N, Okada Y, Yoshida M, Sobue G. Alpha-synuclein-positive structures in association with diffuse neurofibrillary tangles with calcification. Neuropathol Appl Neurobiol 2003; 29:280-7. [PMID: 12787325 DOI: 10.1046/j.1365-2990.2003.00470.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
alpha-Synuclein is known to be a major constituent of the Lewy bodies (LBs) in Parkinson's disease (PD) and the neuronal and glial cytoplasmic inclusions (NCIs, GCIs) in multiple system atrophy. alpha-Synuclein-positive inclusions such as LBs, NCIs and GCIs sometimes show colocalization with tau-positive neurofilaments. Studies using alpha-synuclein immunohistochemistry have often found LBs in the amygdala of patients with familial or sporadic Alzheimer's disease (AD), as well as in patients with Down's syndrome and AD. However, no studies have reported alpha-synuclein-positive structures in cases of diffuse neurofibrillary tangles with calcification (DNTC), which is characterized by numerous neurofibrillary tangles (NFTs) throughout the cerebral cortex but few, if any, senile plaques. We investigated the distribution of alpha-synuclein-positive structures in two cases of DNTC: a 65-year-old woman (brain weight, 850 g) and a 75-year-old woman (brain weight, 800 g). In both cases, severe cerebral atrophy predominant in the temporal lobe was noted. Microscopically, alpha-synuclein-positive intracytoplasmic inclusions and neurites were found in the superior temporal lobe (within the temporal pole), amygdala, parahippocampus, entorhinal cortex and insula, the regions most affected by the NFTs. alpha-Synuclein-positive intracytoplasmic inclusions were rare or absent in other regions of the cerebral cortex and brainstem. This distribution pattern differs from that of PD or dementia with LBs. Our findings suggest that the accumulation pattern of alpha-synuclein is a pathological feature of DNTC, and that DNTC is associated with accumulation of both tau and alpha-synuclein.
Collapse
Affiliation(s)
- N Hishikawa
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Shibuya K, Uchihara T, Nakamura A, Ishiyama M, Yamaoka K, Yagishita S, Iwabuchi K, Kosaka K. Reversible conformational change of tau2 epitope on exposure to detergent in glial cytoplasmic inclusions of multiple system atrophy. Acta Neuropathol 2003; 105:508-14. [PMID: 12677452 DOI: 10.1007/s00401-003-0675-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2002] [Revised: 12/18/2002] [Accepted: 12/18/2002] [Indexed: 10/25/2022]
Abstract
Tau-like immunoreactivity (IR) on glial cytoplasmic inclusions (GCIs) of multiple system atrophy (MSA) was investigated with a panel of anti-tau antibodies and we found that tau2, one of the phosphorylation-independent antibodies, preferentially immunolabeled GCIs. Co-presence (0.03%) of polyethyleneglycol- p-isooctylphenyl ether (Triton X-100, TX) with tau2, however, abolished this IR on GCIs, but did not abolish tau2 IR on neurofibrillary tangles (NFTs). Tau2-immunoreactive bands on immunoblot of brain homogenates from MSA brains were retrieved mainly in a TRIS-saline-soluble fraction, as reported in normal brains. This was in contrast to SDS-soluble fractions from brain with Down's syndrome, which contained tau2-immunoreactive bands of higher molecular weight. It indicates that the appearance of tau2 IR on GCIs is not related to hyperphosphorylation of tau. These tau2-immunoreactive bands, except those from bovine brain, were similarly abolished in the presence of TX (0.06%), and repeated washing after exposure to TX restored the tau2 IR on immunohistochemistry and on immunoblot. These findings can be explained if the modified tau2 epitope undergoes a reversible conformational change on exposure to TX, which is reversible after washing. Because the conformation centered at Ser101 of bovine tau is crucial for its affinity to tau2, the Ser-like conformation mimicked by its human counterpart Pro may represent pathological modification of tau shared by GCIs and NFTs. The relative resistance of tau2 epitope on NFTs on exposure to TX suggests that tau woven into NFTs confers additional stability to the pathological conformation of tau2 epitope. The conformation of the tau2 epitope in GCIs is not as stable as in NFTs, suggesting that tau proteins are not the principal constituents of the fibrillary structures of GCIs, even though they were immunodecorated with tau2. The difference in the susceptibility of the tau2 epitope to TX may distinguish its conformational states, which are variously represented according to disease conditions.
Collapse
Affiliation(s)
- Katsuhiko Shibuya
- Department of Neurology and Psychiatry, Kanagawa Rehabilitation Center, 516 Nanasawa Atsugi-shi, 243-0121 Kanagawa-ken, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Ishizawa T, Mattila P, Davies P, Wang D, Dickson DW. Colocalization of tau and alpha-synuclein epitopes in Lewy bodies. J Neuropathol Exp Neurol 2003; 62:389-97. [PMID: 12722831 DOI: 10.1093/jnen/62.4.389] [Citation(s) in RCA: 279] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The major protein constituent of Lewy bodies (LBs), the pathological hallmark of Parkinson disease and dementia with Lewy bodies, is considered to be alpha-synuclein, but other proteins, in particular the microtubule-associated protein tau, have been implicated in the pathogenesis of LBs. Tau is the major structural component of neurofibrillary tangles (NFTs). Both direct immunochemical studies of partially purified LBs and indirect immunohistochemical studies have suggested that LBs may contain tau, but most of these studies were based upon a single tau antibody, and immunologic cross-reactivity was not completely excluded. To gain insight into the relation between tau and alpha-synuclein in LBs, double immunostaining was performed in Lewy body cases with a rabbit polyclonal antibody to alpha-synuclein and a panel of monoclonal antibodies to phospho- and nonphospho-tau epitopes (Alz50, CP9, CP13, PG5, TG3, PHFI) that spanned the length of the tau molecule. Tau-immunoreactive LBs were present in the medulla in 80% of the cases, irrespective of Braak stage. All tau antibodies recognized at least some LBs, arguing against nonspecific antibody cross-reactivity. In most lesions the tau immunostaining was present at the periphery of the LB. The phospho-tau antibody, TG3, detected more LBs than any of the other tau antibodies. The proportion of LBs with tau immunoreactivity was greatest in neurons vulnerable to NETs, such as those in the locus ceruleus and basal nucleus of Meynert, and least in neurons resistant to NFTs, such as the dorsal motor nucleus of the vagus in the medulla. The present results suggest that tau may coaggregate with alpha-synuclein in LBs, especially in neuronal populations vulnerable to both NFTs and LBs.
Collapse
Affiliation(s)
- Takashi Ishizawa
- Department of Pathology (Neuropathology), Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | | | | | | |
Collapse
|
50
|
Bussell R, Eliezer D. Residual structure and dynamics in Parkinson's disease-associated mutants of alpha-synuclein. J Biol Chem 2001; 276:45996-6003. [PMID: 11590151 DOI: 10.1074/jbc.m106777200] [Citation(s) in RCA: 197] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
alpha-Synuclein (alpha S) is a pre-synaptic protein that has been implicated as a possible causative agent in the pathogenesis of Parkinson's disease (PD). Two autosomal dominant missense mutations in the alpha S gene are associated with early onset PD. Because alpha S is found in an aggregated fibrillar form in the Lewy body deposits characteristic of Parkinson's patients, aggregation of the protein is believed to be related to its involvement in the disease process. The wild type (WT) and early onset mutants A30P and A53T display diverse in vitro aggregation kinetics even though the gross physicochemical and morphological properties of the mutants are highly similar. We used high resolution solution NMR spectroscopy to compare the structural and dynamic properties of the A53T and A30P mutants with those of WT alpha S in the free state. We found that the A30P mutation disrupts a region of residual helical structure that exists in the WT protein, whereas the A53T mutation results in a slight enhancement of a small region around the site of mutation with a preference for extended conformations. Based on these results and on the anticipated effects of these mutations on elements of secondary structure, we proposed a model of how these two PD-linked mutations influence alpha S fibril formation that is consistent with the documented differences in the fibrillization kinetics of the two mutants.
Collapse
Affiliation(s)
- R Bussell
- Department of Physiology, Biophysics, and Molecular Medicine Weill Medical College of Cornell University, New York, New York 10021, USA
| | | |
Collapse
|