1
|
Huang M, McEwan WA. Sensitive detection and propagation of brain-derived tau assemblies in HEK293 based wild-type tau seeding assays. J Biol Chem 2025:108245. [PMID: 39880096 DOI: 10.1016/j.jbc.2025.108245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/28/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
The assembly of tau into filaments defines tauopathies, a group of neurodegenerative diseases including Alzheimer's disease (AD), Pick's disease (PiD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP). The seeded aggregation of tau has been modelled in cell culture using pro-aggregant modifications such as truncation of N- and C-termini and point-mutations within the microtubule-binding repeat domain. This limits the applicability of research findings to sporadic disease, where aggregates contain wild-type, full-length tau. We aimed to develop sensitive and specific biosensor assays for brain-derived tau species utilizing 0N3R and 0N4R tau expressed in HEK293 cells. We further generate a cell line that propagates insoluble tau which is hyperphosphorylated at disease relevant sites and retains a seeding profile similar to AD. We propose these systems as an advance over existing cell-based seeding assays, as they display specificity to the conformation and isoform composition of sporadic human disease.
Collapse
Affiliation(s)
- Melissa Huang
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge, CB2 0AH, United Kingdom.
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge, CB2 0AH, United Kingdom.
| |
Collapse
|
2
|
Marvian AT, Strauss T, Tang Q, Tuck BJ, Keeling S, Rüdiger D, Mirzazadeh Dizaji N, Mohammad-Beigi H, Nuscher B, Chakraborty P, Sutherland DS, McEwan WA, Köglsperger T, Zahler S, Zweckstetter M, Lichtenthaler SF, Wurst W, Schwarz S, Höglinger G. Distinct regulation of Tau Monomer and aggregate uptake and intracellular accumulation in human neurons. Mol Neurodegener 2024; 19:100. [PMID: 39736627 DOI: 10.1186/s13024-024-00786-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND The prion-like spreading of Tau pathology is the leading cause of disease progression in various tauopathies. A critical step in propagating pathologic Tau in the brain is the transport from the extracellular environment and accumulation inside naïve neurons. Current research indicates that human neurons internalize both the physiological extracellular Tau (eTau) monomers and the pathological eTau aggregates. However, similarities or differences in neuronal transport mechanisms between Tau species remain elusive. METHOD Monomers, oligomers, and fibrils of recombinant 2N4R Tau were produced and characterized by biochemical and biophysical methods. A neuronal eTau uptake and accumulation assay was developed for human induced pluripotent stem cell-derived neurons (iPSCNs) and Lund human mesencephalic cells (LUHMES)-derived neurons. Mechanisms of uptake and cellular accumulation of eTau species were studied by using small molecule inhibitors of endocytic mechanisms and siRNAs targeting Tau uptake mediators. RESULTS Extracellular Tau aggregates accumulated more than monomers in human neurons, mainly due to the higher efficiency of small fibrillar and soluble oligomeric aggregates in intraneuronal accumulation. A competition assay revealed a distinction in the neuronal accumulation between physiological eTau Monomers and pathology-relevant aggregates, suggesting differential transport mechanisms. Blocking heparan sulfate proteoglycans (HSPGs) with heparin only inhibited the accumulation of eTau aggregates, whereas monomers' uptake remained unaltered. At the molecular level, the downregulation of genes involved in HSPG synthesis exclusively blocked neuronal accumulation of eTau aggregates but not monomers, suggesting its role in the transport of pathologic Tau. Moreover, the knockdown of LRP1, as a receptor of Tau, mainly reduced the accumulation of monomeric form, confirming its involvement in Tau's physiological transport. CONCLUSION These data propose that despite the similarity in the cellular mechanism, the uptake and accumulation of eTau Monomers and aggregates in human neurons are regulated by different molecular mediators. Thus, they address the possibility of targeting the pathological spreading of Tau aggregates without disturbing the probable physiological or non-pathogenic transport of Tau Monomers.
Collapse
Affiliation(s)
- Amir T Marvian
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany.
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Tabea Strauss
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
| | - Qilin Tang
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
| | - Benjamin J Tuck
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Sophie Keeling
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel Rüdiger
- Department of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Negar Mirzazadeh Dizaji
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Hossein Mohammad-Beigi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs., Lyngby, Denmark
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000, Aarhus C, Denmark
| | - Brigitte Nuscher
- Division of Metabolic Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Pijush Chakraborty
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Gӧttingen, Germany
| | - Duncan S Sutherland
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000, Aarhus C, Denmark
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Thomas Köglsperger
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany
| | - Stefan Zahler
- Department of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Markus Zweckstetter
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Gӧttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Gӧttingen, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences, Technical University Munich, Freising, Germany
| | - Sigrid Schwarz
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Haag, Geriatric Clinic Haag, Oberbayern, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany.
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Department of Neurology, Hanover Medical School, Hanover, Germany.
- Center for Systems Neuroscience, Hanover, Germany.
| |
Collapse
|
3
|
Ijezie EC, Miller MJ, Hardy C, Jarvis AR, Czajka TF, D'Brant L, Rugenstein N, Waickman A, Murphy E, Butler DC. HSV-1 Infection Alters MAPT Splicing and Promotes Tau Pathology in Neural Models of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618683. [PMID: 39464083 PMCID: PMC11507845 DOI: 10.1101/2024.10.16.618683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
INTRODUCTION Herpes simplex virus 1 (HSV-1) infection alters critical markers of Alzheimer's Disease (AD) in neurons. One key marker of AD is the hyperphosphorylation of Tau, accompanied by altered levels of Tau isoforms. However, an imbalance in these Tau splice variants, specifically resulting from altered 3R to 4R MAPT splicing of exon 10, has yet to be directly associated with HSV-1 infection. METHODS To this end, we infected 2D and 3D human neural models with HSV-1 and monitored MAPT splicing and Tau phosphorylation. Further, we transduced SH-SY5Y-neurons with HSV-1 ICP27 which alters RNA splicing to analyze if ICP27 alone is sufficient to induce altered MAPT exon 10 splicing. RESULTS We show that HSV-1 infection induces altered splicing of MAPT exon 10, increasing 4R-Tau protein levels, Tau hyperphosphorylation, and Tau oligomerization. DISCUSSION Our experiments reveal a novel link between HSV-1 infection and the development of cytopathic phenotypes linked with AD progression. HIGHLIGHTS HSV-1 infection in forebrain organoids reduces the neurite length of MAP2-positive neurons.HSV-1 infection increases Tau hyperphosphorylation in both two-month-old and four-month-old forebrain organoids. HSV-1 infection increases Exon 10 containing (4R) MAPT mRNA and 4R-Tau protein expression in both forebrain organoids and human SH-SY5Y-neurons. HSV-1 ICP27 is both necessary and sufficient to induce increased 4R MAPT mRNA and 4R-Tau protein expression in SH-SY5Y-neurons. HSV-1 infection increases Tau oligomerization in both forebrain organoids and SH-SY5Y-neurons.
Collapse
|
4
|
Bhopatkar AA, Bhatt N, Haque MA, Xavier R, Fung L, Jerez C, Kayed R. MAPT mutations associated with familial tauopathies lead to formation of conformationally distinct oligomers that have cross-seeding ability. Protein Sci 2024; 33:e5099. [PMID: 39145409 PMCID: PMC11325167 DOI: 10.1002/pro.5099] [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: 04/21/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 08/16/2024]
Abstract
The microtubule associated protein, tau, is implicated in a multitude of neurodegenerative disorders that are collectively termed as tauopathies. These disorders are characterized by the presence of tau aggregates within the brain of afflicted individuals. Mutations within the MAPT gene that encodes the tau protein form the genetic backdrop for familial forms of tauopathies, such as frontotemporal dementia (FTD), but the molecular consequences of such alterations and their pathological effects are unclear. We sought to investigate the conformational properties of the aggregates of three tau mutants: A152T, P301L, and R406W, all implicated within FTD, and compare them to those of the native form (WT-Tau 2N4R). Our immunochemical analysis reveals that mutants and WT tau oligomers exhibit similar affinity for conformation-specific antibodies but have distinct morphology and secondary structure. Additionally, these oligomers possess different dye-binding properties and varying sensitivity to proteolytic processing. These results point to conformational variety among them. We then tested the ability of the mutant oligomers to cross-seed the aggregation of WT tau monomer. Using similar array of experiments, we found that cross-seeding with mutant aggregates leads to the formation of conformationally unique WT oligomers. The results discussed in this paper provide a novel perspective on the structural properties of oligomeric forms of WT tau 2N4R and its mutant, along with shedding some light on their cross-seeding behavior.
Collapse
Affiliation(s)
- Anukool A. Bhopatkar
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
- Present address:
Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Nemil Bhatt
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Md Anzarul Haque
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rhea Xavier
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Leiana Fung
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
- Present address:
Neuroscience Graduate Program, UT Southwestern Medical CenterDallasTexasUSA
| | - Cynthia Jerez
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
5
|
Ayers J, Lopez TP, Steele IT, Oehler A, Roman-Albarran R, Cleveland E, Chong A, Carlson GA, Condello C, Prusiner SB. Severe neurodegeneration in brains of transgenic rats producing human tau prions. Acta Neuropathol 2024; 148:25. [PMID: 39160375 PMCID: PMC11333523 DOI: 10.1007/s00401-024-02771-5] [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: 04/09/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024]
Abstract
Both wild-type and mutant tau proteins can misfold into prions and self-propagate in the central nervous system of animals and people. To extend the work of others, we investigated the molecular basis of tau prion-mediated neurodegeneration in transgenic (Tg) rats expressing mutant human tau (P301S); this line of Tg rats is denoted Tg12099. We used the rat Prnp promoter to drive the overexpression of mutant tau (P301S) in the human 0N4R isoform. In Tg12099(+/+) rats homozygous for the transgene, ubiquitous expression of mutant human tau resulted in the progressive accumulation of phosphorylated tau inclusions, including silver-positive tangles in the frontal cortices and limbic system. Signs of central nervous system dysfunction were found in terminal Tg12099(+/+) rats exhibiting severe neurodegeneration and profound atrophy of the amygdala and piriform cortex. The greatest increases in tau prion activity were found in the corticolimbic structures. In contrast to the homozygous Tg12099(+/+) rats, we found lower levels of mutant tau in the hemizygous rats, resulting in few neuropathologic changes up to 2 years of age. Notably, these hemizygous rats could be infected by intracerebral inoculation with recombinant tau fibrils or precipitated tau prions from the brain homogenates of sick, aged homozygous Tg12099(+/+) rats. Our studies argue that the regional propagation of tau prions and neurodegeneration in the Tg12099 rats resembles that found in human primary tauopathies. These findings seem likely to advance our understanding of human tauopathies and may lead to effective therapeutics for Alzheimer's disease and other tau prion disorders.
Collapse
Affiliation(s)
- Jacob Ayers
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - T Peter Lopez
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Ian T Steele
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Abby Oehler
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Rigo Roman-Albarran
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Elisa Cleveland
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Alex Chong
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - George A Carlson
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Carlo Condello
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
6
|
Rabanal-Ruiz Y, Pedrero-Prieto CM, Sanchez-Rodriguez L, Flores-Cuadrado A, Saiz-Sanchez D, Frontinan-Rubio J, Ubeda-Banon I, Duran Prado M, Martinez-Marcos A, Peinado JR. Differential accumulation of human β-amyloid and tau from enriched extracts in neuronal and endothelial cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167204. [PMID: 38679217 DOI: 10.1016/j.bbadis.2024.167204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
While Aβ and Tau cellular distribution has been largely studied, the comparative internalization and subcellular accumulation of Tau and Aβ isolated from human brain extracts in endothelial and neuronal cells has not yet been unveiled. We have previously demonstrated that controlled enrichment of Aβ from human brain extracts constitutes a valuable tool to monitor cellular internalization in vitro and in vivo. Herein, we establish an alternative method to strongly enrich Aβ and Tau aggregates from human AD brains, which has allowed us to study and compare the cellular internalization, distribution and toxicity of both proteins within brain barrier endothelial (bEnd.3) and neuronal (Neuro2A) cells. Our findings demonstrate the suitability of human enriched brain extracts to monitor the intracellular distribution of human Aβ and Tau, which, once internalized, show dissimilar sorting to different organelles within the cell and differential toxicity, exhibiting higher toxic effects on neuronal cells than on endothelial cells. While tau is strongly concentrated preferentially in mitochondria, Aβ is distributed predominantly within the endolysosomal system in endothelial cells, whereas the endoplasmic reticulum was its preferential location in neurons. Altogether, our findings display a picture of the interactions that human Aβ and Tau might establish in these cells.
Collapse
Affiliation(s)
- Y Rabanal-Ruiz
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - C M Pedrero-Prieto
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - L Sanchez-Rodriguez
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Flores-Cuadrado
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - D Saiz-Sanchez
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - J Frontinan-Rubio
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - I Ubeda-Banon
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - M Duran Prado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Martinez-Marcos
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Juan R Peinado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
7
|
Mukherjee S, Poudyal M, Dave K, Kadu P, Maji SK. Protein misfolding and amyloid nucleation through liquid-liquid phase separation. Chem Soc Rev 2024; 53:4976-5013. [PMID: 38597222 DOI: 10.1039/d3cs01065a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Liquid-liquid phase separation (LLPS) is an emerging phenomenon in cell physiology and diseases. The weak multivalent interaction prerequisite for LLPS is believed to be facilitated through intrinsically disordered regions, which are prevalent in neurodegenerative disease-associated proteins. These aggregation-prone proteins also exhibit an inherent property for phase separation, resulting in protein-rich liquid-like droplets. The very high local protein concentration in the water-deficient confined microenvironment not only drives the viscoelastic transition from the liquid to solid-like state but also most often nucleate amyloid fibril formation. Indeed, protein misfolding, oligomerization, and amyloid aggregation are observed to be initiated from the LLPS of various neurodegeneration-related proteins. Moreover, in these cases, neurodegeneration-promoting genetic and environmental factors play a direct role in amyloid aggregation preceded by the phase separation. These cumulative recent observations ignite the possibility of LLPS being a prominent nucleation mechanism associated with aberrant protein aggregation. The present review elaborates on the nucleation mechanism of the amyloid aggregation pathway and the possible early molecular events associated with amyloid-related protein phase separation. It also summarizes the recent advancement in understanding the aberrant phase transition of major proteins contributing to neurodegeneration focusing on the common disease-associated factors. Overall, this review proposes a generic LLPS-mediated multistep nucleation mechanism for amyloid aggregation and its implication in neurodegeneration.
Collapse
Affiliation(s)
- Semanti Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Manisha Poudyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Kritika Dave
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
8
|
Arar S, Haque MA, Bhatt N, Zhao Y, Kayed R. Effect of Natural Osmolytes on Recombinant Tau Monomer: Propensity of Oligomerization and Aggregation. ACS Chem Neurosci 2024; 15:1366-1377. [PMID: 38503425 PMCID: PMC10995947 DOI: 10.1021/acschemneuro.3c00614] [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: 09/22/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
The pathological misfolding and aggregation of the microtubule associated protein tau (MAPT), a full length Tau2N4R with 441aa, is considered the principal disease relevant constituent in tauopathies including Alzheimer's disease (AD) with an imbalanced ratio in 3R/4R isoforms. The exact cellular fluid composition, properties, and changes that coincide with tau misfolding, seed formation, and propagation events remain obscure. The proteostasis network, along with the associated osmolytes, is responsible for maintaining the presence of tau in its native structure or dealing with misfolding. In this study, for the first time, the roles of natural brain osmolytes are being investigated for their potential effects on regulating the conformational stability of the tau monomer (tauM) and its propensity to aggregate or disaggregate. Herein, the effects of physiological osmolytes myo-inositol, taurine, trimethyl amine oxide (TMAO), betaine, sorbitol, glycerophosphocholine (GPC), and citrulline on tau's aggregation state were investigated. The overall results indicate the ability of sorbitol and GPC to maintain the monomeric form and prevent aggregation of tau, whereas myo-inositol, taurine, TMAO, betaine, and citrulline promote tau aggregation to different degrees, as revealed by protein morphology in atomic force microscopy images. Biochemical and biophysical methods also revealed that tau proteins adopt different conformations under the influence of these osmolytes. TauM in the presence of all osmolytes expressed no toxicity when tested by a lactate dehydrogenase assay. Investigating the conformational stability of tau in the presence of osmolytes may provide a better understanding of the complex nature of tau aggregation in AD and the protective and/or chaotropic nature of osmolytes.
Collapse
Affiliation(s)
- Sharif Arar
- Mitchell
Center for Neurodegenerative Diseases, University
of Texas Medical Branch, Galveston, Texas 77555, United States
- Departments
of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department
of Chemistry, School of Science, The University
of Jordan, Amman 11942, Jordan
| | - Md Anzarul Haque
- Mitchell
Center for Neurodegenerative Diseases, University
of Texas Medical Branch, Galveston, Texas 77555, United States
- Departments
of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Nemil Bhatt
- Mitchell
Center for Neurodegenerative Diseases, University
of Texas Medical Branch, Galveston, Texas 77555, United States
- Departments
of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Yingxin Zhao
- Department
of Internal Medicine, University of Texas
Medical Branch, Galveston, Texas 77555, United States
- Institute
for Translational Sciences, University of
Texas Medical Branch, Galveston, Texas 77555, United States
| | - Rakez Kayed
- Mitchell
Center for Neurodegenerative Diseases, University
of Texas Medical Branch, Galveston, Texas 77555, United States
- Departments
of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
9
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
10
|
Xu H, Qiu Q, Hu P, Hoxha K, Jang E, O'Reilly M, Kim C, He Z, Marotta N, Changolkar L, Zhang B, Wu H, Schellenberg GD, Kraemer B, Luk KC, Lee EB, Trojanowski JQ, Brunden KR, Lee VMY. MSUT2 regulates tau spreading via adenosinergic signaling mediated ASAP1 pathway in neurons. Acta Neuropathol 2024; 147:55. [PMID: 38472475 PMCID: PMC10933148 DOI: 10.1007/s00401-024-02703-3] [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: 12/06/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024]
Abstract
Inclusions comprised of microtubule-associated protein tau (tau) are implicated in a group of neurodegenerative diseases, collectively known as tauopathies, that include Alzheimer's disease (AD). The spreading of misfolded tau "seeds" along neuronal networks is thought to play a crucial role in the progression of tau pathology. Consequently, restricting the release or uptake of tau seeds may inhibit the spread of tau pathology and potentially halt the advancement of the disease. Previous studies have demonstrated that the Mammalian Suppressor of Tauopathy 2 (MSUT2), an RNA binding protein, modulates tau pathogenesis in a transgenic mouse model. In this study, we investigated the impact of MSUT2 on tau pathogenesis using tau seeding models. Our findings indicate that the loss of MSUT2 mitigates human tau seed-induced pathology in neuron cultures and mouse models. In addition, MSUT2 regulates many gene transcripts, including the Adenosine Receptor 1 (A1AR), and we show that down regulation or inhibition of A1AR modulates the activity of the "ArfGAP with SH3 Domain, Ankyrin Repeat, and PH Domain 1 protein" (ASAP1), thereby influencing the internalization of pathogenic tau seeds into neurons resulting in reduction of tau pathology.
Collapse
Affiliation(s)
- Hong Xu
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Qi Qiu
- Department of Genetics, Penn Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peng Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education), Shanghai Ocean University, Shanghai, China
| | - Kevt'her Hoxha
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elliot Jang
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mia O'Reilly
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Kim
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhuohao He
- Interdisciplinary Research Center On Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Nicholas Marotta
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lakshmi Changolkar
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bin Zhang
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, Penn Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Kraemer
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kurt R Brunden
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Mamais A, Sanyal A, Fajfer A, Zykoski CG, Guldin M, Riley-DiPaolo A, Subrahmanian N, Gibbs W, Lin S, LaVoie MJ. The LRRK2 kinase substrates RAB8a and RAB10 contribute complementary but distinct disease-relevant phenotypes in human neurons. Stem Cell Reports 2024; 19:163-173. [PMID: 38307024 PMCID: PMC10874859 DOI: 10.1016/j.stemcr.2024.01.001] [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: 08/30/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 02/04/2024] Open
Abstract
Mutations in the LRRK2 gene cause familial Parkinson's disease presenting with pleomorphic neuropathology that can involve α-synuclein or tau accumulation. LRRK2 mutations are thought to converge upon a pathogenic increase in LRRK2 kinase activity. A subset of small RAB GTPases has been identified as LRRK2 substrates, with LRRK2-dependent phosphorylation resulting in RAB inactivation. We used CRISPR-Cas9 genome editing to generate a novel series of isogenic iPSC lines deficient in the two most well-validated LRRK2 substrates, RAB8a and RAB10, from deeply phenotyped healthy control lines. Thorough characterization of NGN2-induced neurons revealed opposing effects of RAB8a and RAB10 deficiency on lysosomal pH and Golgi organization, with isolated effects of RAB8a and RAB10 ablation on α-synuclein and tau, respectively. Our data demonstrate largely antagonistic effects of genetic RAB8a or RAB10 inactivation, which provide discrete insight into the pathologic features of their biochemical inactivation by pathogenic LRRK2 mutation in human disease.
Collapse
Affiliation(s)
- Adamantios Mamais
- Center for Translational Research in Neurodegenerative Disease and Fixel Institute for Neurologic Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Anwesha Sanyal
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Austin Fajfer
- Center for Translational Research in Neurodegenerative Disease and Fixel Institute for Neurologic Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Catherine G Zykoski
- Center for Translational Research in Neurodegenerative Disease and Fixel Institute for Neurologic Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Michael Guldin
- Center for Translational Research in Neurodegenerative Disease and Fixel Institute for Neurologic Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | | | - Nitya Subrahmanian
- Center for Translational Research in Neurodegenerative Disease and Fixel Institute for Neurologic Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Whitney Gibbs
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven Lin
- Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew J LaVoie
- Center for Translational Research in Neurodegenerative Disease and Fixel Institute for Neurologic Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA; Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Guo X, Yan L, Zhang D, Zhao Y. Passive immunotherapy for Alzheimer's disease. Ageing Res Rev 2024; 94:102192. [PMID: 38219962 DOI: 10.1016/j.arr.2024.102192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/03/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease characterized by cognitive impairment with few therapeutic options. Despite many failures in developing AD treatment during the past 20 years, significant advances have been achieved in passive immunotherapy of AD very recently. Here, we review characteristics, clinical trial data, and mechanisms of action for monoclonal antibodies (mAbs) targeting key players in AD pathogenesis, including amyloid-β (Aβ), tau and neuroinflammation modulators. We emphasized the efficacy of lecanemab and donanemab on cognition and amyloid clearance in AD patients in phase III clinical trials and discussed factors that may contribute to the efficacy and side effects of anti-Aβ mAbs. In addition, we provided important information on mAbs targeting tau or inflammatory regulators in clinical trials, and indicated that mAbs against the mid-region of tau or pathogenic tau have therapeutic potential for AD. In conclusion, passive immunotherapy targeting key players in AD pathogenesis offers a promising strategy for effective AD treatment.
Collapse
Affiliation(s)
- Xiaoyi Guo
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Li Yan
- School of Traditional Chinese Medicine, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong 510632, China
| | - Denghong Zhang
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Yingjun Zhao
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
13
|
Creekmore BC, Watanabe R, Lee EB. Neurodegenerative Disease Tauopathies. ANNUAL REVIEW OF PATHOLOGY 2024; 19:345-370. [PMID: 37832941 PMCID: PMC11009985 DOI: 10.1146/annurev-pathmechdis-051222-120750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Tauopathies are a diverse group of progressive and fatal neurodegenerative diseases characterized by aberrant tau inclusions in the central nervous system. Tau protein forms pathologic fibrillar aggregates that are typically closely associated with neuronal cell death, leading to varied clinical phenotypes including dementia, movement disorders, and motor neuron disease. In this review, we describe the clinicopathologic features of tauopathies and highlight recent advances in understanding the mechanisms that lead to spread of pathologic aggregates through interconnected neuronal pathways. The cell-to-cell propagation of tauopathy is then linked to posttranslational modifications, tau fibril structural variants, and the breakdown of cellular protein quality control.
Collapse
Affiliation(s)
- Benjamin C Creekmore
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Ryohei Watanabe
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Edward B Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
14
|
Goedert M, Crowther RA, Scheres SHW, Spillantini MG. Tau and neurodegeneration. Cytoskeleton (Hoboken) 2024; 81:95-102. [PMID: 38073060 PMCID: PMC7615684 DOI: 10.1002/cm.21812] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 01/24/2024]
Abstract
First identified in 1975, tau was implicated in Alzheimer's disease 10 years later. Filamentous tangle inclusions were known to be made of hyperphosphorylated tau by 1991, with similar inclusions gaining recognition for being associated with other neurodegenerative diseases. In 1998, mutations in MAPT, the gene that encodes tau, were identified as the cause of a dominantly inherited form of frontotemporal dementia with abundant filamentous tau inclusions. While this result indicated that assembly of tau into aberrant filaments is sufficient to drive neurodegeneration and dementia, most cases of tauopathy are sporadic. More recent work in experimental systems showed that filamentous assemblies of tau may first form in one brain area, and then spread to others in a prion-like fashion. Beginning in 2017, work on human brains using high-resolution techniques has led to a structure-based classification of tauopathies, which has opened the door to a better understanding of the significance of tau filament formation.
Collapse
Affiliation(s)
- Michel Goedert
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK
| | - R. Anthony Crowther
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK
| | - Sjors H. W. Scheres
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK
| | | |
Collapse
|
15
|
Lövestam S, Li D, Wagstaff JL, Kotecha A, Kimanius D, McLaughlin SH, Murzin AG, Freund SMV, Goedert M, Scheres SHW. Disease-specific tau filaments assemble via polymorphic intermediates. Nature 2024; 625:119-125. [PMID: 38030728 PMCID: PMC10764278 DOI: 10.1038/s41586-023-06788-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Intermediate species in the assembly of amyloid filaments are believed to play a central role in neurodegenerative diseases and may constitute important targets for therapeutic intervention1,2. However, structural information about intermediate species has been scarce and the molecular mechanisms by which amyloids assemble remain largely unknown. Here we use time-resolved cryogenic electron microscopy to study the in vitro assembly of recombinant truncated tau (amino acid residues 297-391) into paired helical filaments of Alzheimer's disease or into filaments of chronic traumatic encephalopathy3. We report the formation of a shared first intermediate amyloid filament, with an ordered core comprising residues 302-316. Nuclear magnetic resonance indicates that the same residues adopt rigid, β-strand-like conformations in monomeric tau. At later time points, the first intermediate amyloid disappears and we observe many different intermediate amyloid filaments, with structures that depend on the reaction conditions. At the end of both assembly reactions, most intermediate amyloids disappear and filaments with the same ordered cores as those from human brains remain. Our results provide structural insights into the processes of primary and secondary nucleation of amyloid assembly, with implications for the design of new therapies.
Collapse
Affiliation(s)
| | - David Li
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | - Abhay Kotecha
- Thermo Fisher Scientific, Eindhoven, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
16
|
Neațu M, Covaliu A, Ioniță I, Jugurt A, Davidescu EI, Popescu BO. Monoclonal Antibody Therapy in Alzheimer's Disease. Pharmaceutics 2023; 16:60. [PMID: 38258071 PMCID: PMC11154277 DOI: 10.3390/pharmaceutics16010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative condition marked by the progressive deterioration of cognitive abilities, memory impairment, and the accumulation of abnormal proteins, specifically beta-amyloid plaques and tau tangles, within the brain. Despite extensive research efforts, Alzheimer's disease remains without a cure, presenting a significant global healthcare challenge. Recently, there has been an increased focus on antibody-based treatments as a potentially effective method for dealing with Alzheimer's disease. This paper offers a comprehensive overview of the current status of research on antibody-based molecules as therapies for Alzheimer's disease. We will briefly mention their mechanisms of action, therapeutic efficacy, and safety profiles while addressing the challenges and limitations encountered during their development. We also highlight some crucial considerations in antibody-based treatment development, including patient selection criteria, dosing regimens, or safety concerns. In conclusion, antibody-based therapies present a hopeful outlook for addressing Alzheimer's disease. While challenges remain, the accumulating evidence suggests that these therapies may offer substantial promise in ameliorating or preventing the progression of this debilitating condition, thus potentially enhancing the quality of life for the millions of individuals and families affected by Alzheimer's disease worldwide.
Collapse
Affiliation(s)
- Monica Neațu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Anca Covaliu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Iulia Ioniță
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Ana Jugurt
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Eugenia Irene Davidescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Department of Cell Biology, Neurosciences and Experimental Myology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
17
|
Shulman M, Kong J, O'Gorman J, Ratti E, Rajagovindan R, Viollet L, Huang E, Sharma S, Racine AM, Czerkowicz J, Graham D, Li Y, Hering H, Haeberlein SB. TANGO: a placebo-controlled randomized phase 2 study of efficacy and safety of the anti-tau monoclonal antibody gosuranemab in early Alzheimer's disease. NATURE AGING 2023; 3:1591-1601. [PMID: 38012285 PMCID: PMC10724064 DOI: 10.1038/s43587-023-00523-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/10/2023] [Indexed: 11/29/2023]
Abstract
In Alzheimer's disease, the spread of aberrantly phosphorylated tau is an important criterion in the Braak staging of disease severity and correlates with disease symptomatology. Here, we report the results of TANGO ( NCT03352557 ), a randomized, double-blind, placebo-controlled, parallel-group and multiple-dose long-term trial of gosuranemab-a monoclonal antibody to N-terminal tau-in patients with early Alzheimer's disease. The primary objective was to assess the safety and tolerability of gosuranemab compared to placebo. The secondary objectives were to assess the efficacy of multiple doses of gosuranemab in slowing cognitive and functional impairment (using the Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) scores at week 78) and evaluate the immunogenicity of gosuranemab (using the incidence of anti-gosuranemab antibody responses). Participants were randomized (n = 654); received (n = 650) low-dose (125 mg once every 4 weeks (q4w), n = 58; 375 mg q12w, n = 58), intermediate-dose (600 mg q4w, n = 106) or high-dose (2,000 mg q4w, n = 214) gosuranemab or placebo (q4w, n = 214) intravenously for 78 weeks; and assigned to cerebrospinal fluid (n = 327) and/or tau positron emission tomography (n = 357) biomarker substudies. Gosuranemab had an acceptable safety profile and was generally well tolerated (incidence of serious adverse events: placebo, 12.1%; low dose, 10.3%; intermediate dose, 12.3%; high dose, 11.7%). The incidence of treatment-emergent gosuranemab antibody responses was low at all time points. No significant effects were identified in cognitive and functional tests as no dose resulted in a favorable change from the baseline CDR-SB score at week 78 compared to placebo control (adjusted mean change: placebo, 1.85; low dose, 2.20; intermediate dose, 2.24; high dose, 1.85). At week 76, all doses caused significant (P < 0.0001) reductions in the cerebrospinal fluid levels of unbound N-terminal tau compared to placebo.
Collapse
Affiliation(s)
| | | | | | - Elena Ratti
- Biogen, Cambridge, MA, USA
- Takeda Pharmaceuticals, Cambridge, MA, USA
| | | | - Louis Viollet
- Biogen, Cambridge, MA, USA
- Moderna, Cambridge, MA, USA
| | | | | | - Annie M Racine
- Biogen, Cambridge, MA, USA
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
18
|
Steinebrei M, Baur J, Pradhan A, Kupfer N, Wiese S, Hegenbart U, Schönland SO, Schmidt M, Fändrich M. Common transthyretin-derived amyloid fibril structures in patients with hereditary ATTR amyloidosis. Nat Commun 2023; 14:7623. [PMID: 37993462 PMCID: PMC10665346 DOI: 10.1038/s41467-023-43301-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023] Open
Abstract
Systemic ATTR amyloidosis is an increasingly important protein misfolding disease that is provoked by the formation of amyloid fibrils from transthyretin protein. The pathological and clinical disease manifestations and the number of pathogenic mutational changes in transthyretin are highly diverse, raising the question whether the different mutations may lead to different fibril morphologies. Using cryo-electron microscopy, however, we show here that the fibril structure is remarkably similar in patients that are affected by different mutations. Our data suggest that the circumstances under which these fibrils are formed and deposited inside the body - and not only the fibril morphology - are crucial for defining the phenotypic variability in many patients.
Collapse
Affiliation(s)
- Maximilian Steinebrei
- Institute of Protein Biochemistry, Ulm University, Helmholtzstrasse 8/1, Ulm, D-89081, Germany.
| | - Julian Baur
- Institute of Protein Biochemistry, Ulm University, Helmholtzstrasse 8/1, Ulm, D-89081, Germany
| | - Anaviggha Pradhan
- Institute of Protein Biochemistry, Ulm University, Helmholtzstrasse 8/1, Ulm, D-89081, Germany
| | - Niklas Kupfer
- Institute of Protein Biochemistry, Ulm University, Helmholtzstrasse 8/1, Ulm, D-89081, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Medical Faculty, Ulm University, Ulm, D-89081, Germany
| | - Ute Hegenbart
- Medical Department V, Amyloidosis Center, Heidelberg, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, D-69120, Germany
| | - Stefan O Schönland
- Medical Department V, Amyloidosis Center, Heidelberg, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, D-69120, Germany
| | - Matthias Schmidt
- Institute of Protein Biochemistry, Ulm University, Helmholtzstrasse 8/1, Ulm, D-89081, Germany
| | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, Helmholtzstrasse 8/1, Ulm, D-89081, Germany
| |
Collapse
|
19
|
Schweighauser M, Murzin AG, Macdonald J, Lavenir I, Crowther RA, Scheres SHW, Goedert M. Cryo-EM structures of tau filaments from the brains of mice transgenic for human mutant P301S Tau. Acta Neuropathol Commun 2023; 11:160. [PMID: 37798679 PMCID: PMC10552433 DOI: 10.1186/s40478-023-01658-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023] Open
Abstract
Mice transgenic for human mutant P301S tau are widely used as models for human tauopathies. They develop neurodegeneration and abundant filamentous inclusions made of human mutant four-repeat tau. Here we used electron cryo-microscopy (cryo-EM) to determine the structures of tau filaments from the brains of Tg2541 and PS19 mice. Both lines express human P301S tau (0N4R for Tg2541 and 1N4R for PS19) on mixed genetic backgrounds and downstream of different promoters (murine Thy1 for Tg2541 and murine Prnp for PS19). The structures of tau filaments from Tg2541 and PS19 mice differ from each other and those of wild-type tau filaments from human brains. The structures of tau filaments from the brains of humans with mutations P301L, P301S or P301T in MAPT are not known. Filaments from the brains of Tg2541 and PS19 mice share a substructure at the junction of repeats 2 and 3, which comprises residues I297-V312 of tau and includes the P301S mutation. The filament core from the brainstem of Tg2541 mice consists of residues K274-H329 of tau and two disconnected protein densities. Two non-proteinaceous densities are also in evidence. The filament core from the cerebral cortex of line PS19 extends from residues G271-P364 of tau. One strong non-proteinaceous density is also present. Unlike the tau filaments from human brains, the sequences following repeat 4 are missing from the cores of tau filaments from the brains of Tg2541 and PS19 mice.
Collapse
Affiliation(s)
| | - Alexey G Murzin
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | | - Isabelle Lavenir
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | | - Sjors H W Scheres
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| | - Michel Goedert
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
20
|
Ahn S, Suh JS, Jang YK, Kim H, Han K, Lee Y, Choi G, Kim TJ. TAUCON and TAUCOM: A novel biosensor based on fluorescence resonance energy transfer for detecting tau hyperphosphorylation-associated cellular pathologies. Biosens Bioelectron 2023; 237:115533. [PMID: 37517333 DOI: 10.1016/j.bios.2023.115533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/02/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023]
Abstract
Tauopathies are neurodegenerative diseases characterized by abnormal conformational changes in tau protein. Early hyperphosphorylation-induced conformational changes are considered a hallmark of tauopathy, but real-time tracking methods are lacking. Here, we present two novel fluorescence resonance energy transfer (FRET)-based tau biosensors that detect such changes with high spatiotemporal resolution at the single-cell level. The TAUCON biosensor measures instantaneous conformational changes in hyperphosphorylated tau within 20 min, while the TAUCOM biosensor detects changes in the paper-clip structure of microtubule-associated tau. Our biosensors provide faster and more precise detection than conventional methods and can serve as valuable tools for investigating the initial causes, mechanisms, progression, and treatment of tauopathies.
Collapse
Affiliation(s)
- Sanghyun Ahn
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jung-Soo Suh
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Yoon-Kwan Jang
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Heonsu Kim
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Kiseok Han
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Yerim Lee
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Gyuho Choi
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea
| | - Tae-Jin Kim
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea; Department of Biological Sciences, College of Natural Sciences, Pusan National University, Pusan, 46241, Republic of Korea; Institute of System Biology, Pusan National University, Pusan, 46241, Republic of Korea.
| |
Collapse
|
21
|
Hromadkova L, Kim C, Haldiman T, Peng L, Zhu X, Cohen M, de Silva R, Safar JG. Evolving prion-like tau conformers differentially alter postsynaptic proteins in neurons inoculated with distinct isolates of Alzheimer's disease tau. Cell Biosci 2023; 13:174. [PMID: 37723591 PMCID: PMC10507869 DOI: 10.1186/s13578-023-01133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023] Open
Abstract
OBJECTIVES Although accumulation of misfolded tau species has been shown to predict cognitive decline in patients with Alzheimer's disease (AD) and other tauopathies but with the remarkable diversity of clinical manifestations, neuropathology profiles, and time courses of disease progression remaining unexplained by current genetic data. We considered the diversity of misfolded tau conformers present in individual AD cases as an underlying driver of the phenotypic variations of AD and progressive loss of synapses. METHODS To model the mechanism of tau propagation and synaptic toxicity of distinct tau conformers, we inoculated wild-type primary mouse neurons with structurally characterized Sarkosyl-insoluble tau isolates from the frontal cortex of six AD cases and monitored the impact for fourteen days. We analyzed the accumulation rate, tau isoform ratio, and conformational characteristics of de novo-induced tau aggregates with conformationally sensitive immunoassays, and the dynamics of synapse formation, maintenance, and their loss using a panel of pre-and post-synaptic markers. RESULTS At the same concentrations of tau, the different AD tau isolates induced accumulation of misfolded predominantly 4-repeat tau aggregates at different rates in mature neurons, and demonstrated distinct conformational characteristics corresponding to the original AD brain tau. The time-course of the formation of misfolded tau aggregates and colocalization correlated with significant loss of synapses in tau-inoculated cell cultures and the reduction of synaptic connections implicated the disruption of postsynaptic compartment as an early event. CONCLUSIONS The data obtained with mature neurons expressing physiological levels and adult isoforms of tau protein demonstrate markedly different time courses of endogenous tau misfolding and differential patterns of post-synaptic alterations. These and previous biophysical data argue for an ensemble of various misfolded tau aggregates in individual AD brains and template propagation of their homologous conformations in neurons with different rates and primarily postsynaptic interactors. Modeling tau aggregation in mature differentiated neurons provides a platform for investigating divergent molecular mechanisms of tau strain propagation and for identifying common structural features of misfolded tau and critical interactors for new therapeutic targets and approaches in AD.
Collapse
Affiliation(s)
- Lenka Hromadkova
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Chae Kim
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Tracy Haldiman
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Lihua Peng
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Xiongwei Zhu
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
- Departments of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Mark Cohen
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
- National Prion Disease Pathology Surveillance Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Rohan de Silva
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, WC1N 1PJ, UK
| | - Jiri G Safar
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA.
- Departments of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Departments of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
22
|
Scheres SHW, Ryskeldi-Falcon B, Goedert M. Molecular pathology of neurodegenerative diseases by cryo-EM of amyloids. Nature 2023; 621:701-710. [PMID: 37758888 DOI: 10.1038/s41586-023-06437-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 07/14/2023] [Indexed: 09/29/2023]
Abstract
Abnormal assembly of tau, α-synuclein, TDP-43 and amyloid-β proteins into amyloid filaments defines most human neurodegenerative diseases. Genetics provides a direct link between filament formation and the causes of disease. Developments in cryo-electron microscopy (cryo-EM) have made it possible to determine the atomic structures of amyloids from postmortem human brains. Here we review the structures of brain-derived amyloid filaments that have been determined so far and discuss their impact on research into neurodegeneration. Whereas a given protein can adopt many different filament structures, specific amyloid folds define distinct diseases. Amyloid structures thus provide a description of neuropathology at the atomic level and a basis for studying disease. Future research should focus on model systems that replicate the structures observed in disease to better understand the molecular mechanisms of disease and develop improved diagnostics and therapies.
Collapse
Affiliation(s)
- Sjors H W Scheres
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| | | | - Michel Goedert
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
23
|
Flavin WP, Hosseini H, Ruberti JW, Kavehpour HP, Giza CC, Prins ML. Traumatic brain injury and the pathways to cerebral tau accumulation. Front Neurol 2023; 14:1239653. [PMID: 37638180 PMCID: PMC10450935 DOI: 10.3389/fneur.2023.1239653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Tau is a protein that has received national mainstream recognition for its potential negative impact to the brain. This review succinctly provides information on the structure of tau and its normal physiological functions, including in hibernation and changes throughout the estrus cycle. There are many pathways involved in phosphorylating tau including diabetes, stroke, Alzheimer's disease (AD), brain injury, aging, and drug use. The common mechanisms for these processes are put into context with changes observed in mild and repetitive mild traumatic brain injury (TBI). The phosphorylation of tau is a part of the progression to pathology, but the ability for tau to aggregate and propagate is also addressed. Summarizing both the functional and dysfunctional roles of tau can help advance our understanding of this complex protein, improve our care for individuals with a history of TBI, and lead to development of therapeutic interventions to prevent or reverse tau-mediated neurodegeneration.
Collapse
Affiliation(s)
- William P. Flavin
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Helia Hosseini
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
| | - Jeffrey W. Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - H. Pirouz Kavehpour
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Mechanical and Aerospace Engineering, UCLA, Los Angeles, CA, United States
| | - Christopher C. Giza
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Mayumi L. Prins
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| |
Collapse
|
24
|
Celauro L, Burato A, Zattoni M, De Cecco E, Fantuz M, Cazzaniga FA, Bistaffa E, Moda F, Legname G. Different tau fibril types reduce prion level in chronically and de novo infected cells. J Biol Chem 2023; 299:105054. [PMID: 37454740 PMCID: PMC10432985 DOI: 10.1016/j.jbc.2023.105054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/06/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023] Open
Abstract
Neurodegenerative diseases are often characterized by the codeposition of different amyloidogenic proteins, normally defining distinct proteinopathies. An example is represented by prion diseases, where the classical deposition of the aberrant conformational isoform of the prion protein (PrPSc) can be associated with tau insoluble species, which are usually involved in another class of diseases called tauopathies. How this copresence of amyloidogenic proteins can influence the progression of prion diseases is still a matter of debate. Recently, the cellular form of the prion protein, PrPC, has been investigated as a possible receptor of amyloidogenic proteins, since its binding activity with Aβ, tau, and α-synuclein has been reported, and it has been linked to several neurotoxic behaviors exerted by these proteins. We have previously shown that the treatment of chronically prion-infected cells with tau K18 fibrils reduced PrPSc levels. In this work, we further explored this mechanism by using another tau construct that includes the sequence that forms the core of Alzheimer's disease tau filaments in vivo to obtain a distinct fibril type. Despite a difference of six amino acids, these two constructs form fibrils characterized by distinct biochemical and biological features. However, their effects on PrPSc reduction were comparable and probably based on the binding to PrPC at the plasma membrane, inhibiting the pathological conversion event. Our results suggest PrPC as receptor for different types of tau fibrils and point out a role of tau amyloid fibrils in preventing the pathological PrPC to PrPSc conformational change.
Collapse
Affiliation(s)
- Luigi Celauro
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Anna Burato
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Marco Zattoni
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Elena De Cecco
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Marco Fantuz
- Fondazione per la Ricerca Biomedica Avanzata VIMM, Padova, Italy; Dipartimento di Biologia, Università degli Studi di Padova, Padova, Italy
| | - Federico Angelo Cazzaniga
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Edoardo Bistaffa
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabio Moda
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Legname
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy.
| |
Collapse
|
25
|
Dimou E, Katsinelos T, Meisl G, Tuck BJ, Keeling S, Smith AE, Hidari E, Lam JYL, Burke M, Lövestam S, Ranasinghe RT, McEwan WA, Klenerman D. Super-resolution imaging unveils the self-replication of tau aggregates upon seeding. Cell Rep 2023; 42:112725. [PMID: 37393617 PMCID: PMC7614924 DOI: 10.1016/j.celrep.2023.112725] [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: 10/31/2022] [Revised: 04/03/2023] [Accepted: 06/14/2023] [Indexed: 07/04/2023] Open
Abstract
Tau is a soluble protein interacting with tubulin to stabilize microtubules. However, under pathological conditions, it becomes hyperphosphorylated and aggregates, a process that can be induced by treating cells with exogenously added tau fibrils. Here, we employ single-molecule localization microscopy to resolve the aggregate species formed in early stages of seeded tau aggregation. We report that entry of sufficient tau assemblies into the cytosol induces the self-replication of small tau aggregates, with a doubling time of 5 h inside HEK cells and 1 day in murine primary neurons, which then grow into fibrils. Seeding occurs in the vicinity of the microtubule cytoskeleton, is accelerated by the proteasome, and results in release of small assemblies into the media. In the absence of seeding, cells still spontaneously form small aggregates at lower levels. Overall, our work provides a quantitative picture of the early stages of templated seeded tau aggregation in cells.
Collapse
Affiliation(s)
- Eleni Dimou
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK.
| | - Taxiarchis Katsinelos
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Georg Meisl
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Benjamin J Tuck
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - Sophie Keeling
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - Annabel E Smith
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - Eric Hidari
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - Jeff Y L Lam
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - Melanie Burke
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - Sofia Lövestam
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Rohan T Ranasinghe
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - William A McEwan
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Hills Road, Cambridge CB2 0AH, UK.
| |
Collapse
|
26
|
Horie K, Li Y, Barthélemy NR, Gordon BA, Hassenstab J, Benzinger TL, Fagan AM, Morris JC, Karch CM, Xiong C, Allegri R, Mendez PC, Ikeuchi T, Kasuga K, Noble J, Farlow M, Chhatwal J, Day GS, Schofield PR, Masters CL, Levin J, Jucker M, Lee JH, Hoon Roh J, Sato C, Sachdev P, Koyama A, Reyderman L, Bateman RJ, McDade E. Change in Cerebrospinal Fluid Tau Microtubule Binding Region Detects Symptom Onset, Cognitive Decline, Tangles, and Atrophy in Dominantly Inherited Alzheimer's Disease. Ann Neurol 2023; 93:1158-1172. [PMID: 36843330 PMCID: PMC10238659 DOI: 10.1002/ana.26620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/28/2023]
Abstract
OBJECTIVE Identifying cerebrospinal fluid measures of the microtubule binding region of tau (MTBR-tau) species that reflect tau aggregation could provide fluid biomarkers that track Alzheimer's disease related neurofibrillary tau pathological changes. We examined the cerebrospinal fluid (CSF) MTBR-tau species in dominantly inherited Alzheimer's disease (DIAD) mutation carriers to assess the association with Alzheimer's disease (AD) biomarkers and clinical symptoms. METHODS Cross-sectional and longitudinal CSF from 229 DIAD mutation carriers and 130 mutation non-carriers had sequential characterization of N-terminal/mid-domain phosphorylated tau (p-tau) followed by MTBR-tau species and tau positron emission tomography (tau PET), other soluble tau and amyloid biomarkers, comprehensive clinical and cognitive assessments, and brain magnetic resonance imaging of atrophy. RESULTS CSF MTBR-tau species located within the putative "border" region and one species corresponding to the "core" region of aggregates in neurofibrillary tangles (NFTs) increased during the presymptomatic stage and decreased during the symptomatic stage. The "border" MTBR-tau species were associated with amyloid pathology and CSF p-tau; whereas the "core" MTBR-tau species were associated stronger with tau PET and CSF measures of neurodegeneration. The ratio of the border to the core species provided a continuous measure of increasing amounts that tracked clinical progression and NFTs. INTERPRETATION Changes in CSF soluble MTBR-tau species preceded the onset of dementia, tau tangle increase, and atrophy in DIAD. The ratio of 4R-specific MTBR-tau (border) to the NFT (core) MTBR-tau species corresponds to the pathology of NFTs in DIAD and change with disease progression. The dynamics between different MTBR-tau species in the CSF may serve as a marker of tau-related disease progression and target engagement of anti-tau therapeutics. ANN NEUROL 2023;93:1158-1172.
Collapse
Affiliation(s)
- Kanta Horie
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Eisai Inc., Nutley, NJ, 07110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Nicolas R. Barthélemy
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Tammie. L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Ricardo Allegri
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI) Instituto de Investigaciones Neurológicas Raúl Correa, Buenos Aires, Argentina
| | - Patricio Chrem Mendez
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI) Instituto de Investigaciones Neurológicas Raúl Correa, Buenos Aires, Argentina
| | | | | | - James Noble
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, G.H. Sergievsky Center, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - Martin Farlow
- Department of Neurology, Indiana University, Indianapolis, IN 46202, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Harvard Medical School Boston, MA 02114, USA
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL 32224, USA
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, 2031 NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Colin L. Masters
- The Florey Institute and the University of Melbourne, Parkville, Victoria 3010, Australia
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Marchioninistr 15, D-83177 Munchen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, Ludwig-Maximilians Universität München, Marchioninistr 15, 83177 Munich, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE) Tübingen; and Hertie-Institute for Clinical Brain Research, University of Tübingen, D-72076 Tübingen, Germany
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, Seoul 05505, Korea
| | - Jee Hoon Roh
- Departments of Biomedical Sciences, Physiology, and Neurology, Korea University College of Medicine, Seoul 02841, Korea
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | | | | | | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | | |
Collapse
|
27
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
28
|
Mamais A, Sanyal A, Fajfer A, Zykoski CG, Guldin M, Riley-DiPaolo A, Subrahmanian N, Gibbs W, Lin S, LaVoie MJ. The LRRK2 kinase substrates Rab8a and Rab10 contribute complementary but distinct disease-relevant phenotypes in human neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.30.538317. [PMID: 37163109 PMCID: PMC10168414 DOI: 10.1101/2023.04.30.538317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Mutations in the LRRK2 gene cause familial Parkinson's disease presenting with pleomorphic neuropathology that can involve α-synuclein or tau accumulation. LRRK2 mutations are thought to converge toward a pathogenic increase in LRRK2 kinase activity. A subset of small Rab GTPases have been identified as LRRK2 substrates, with LRRK2-dependent phosphorylation resulting in Rab inactivation. We used CRISPR/Cas9 genome editing to generate a novel series of isogenic iPSC lines deficient in the two most well validated LRRK2 substrates, Rab8a and Rab10, from two independent, deeply phenotyped healthy control lines. Thorough characterization of NGN2-induced neurons revealed divergent effects of Rab8a and Rab10 deficiency on lysosomal pH, LAMP1 association with Golgi, α-synuclein insolubility and tau phosphorylation, while parallel effects on lysosomal numbers and Golgi clustering were observed. Our data demonstrate largely antagonistic effects of genetic Rab8a or Rab10 inactivation which provide discrete insight into the pathologic features of their biochemical inactivation by pathogenic LRRK2 mutation.
Collapse
Affiliation(s)
- Adamantios Mamais
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Anwesha Sanyal
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Austin Fajfer
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Catherine G. Zykoski
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Michael Guldin
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Alexis Riley-DiPaolo
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Nitya Subrahmanian
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Whitney Gibbs
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Steven Lin
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
| | - Matthew J. LaVoie
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, USA
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Dong Y, Lu J, Zhang S, Chen L, Wen J, Wang F, Mao Y, Li L, Zhang J, Liao S, Dong L. Design, synthesis and bioevaluation of 1,2,4-thiadiazolidine-3,5-dione derivatives as potential GSK-3β inhibitors for the treatment of Alzheimer's disease. Bioorg Chem 2023; 134:106446. [PMID: 36868127 DOI: 10.1016/j.bioorg.2023.106446] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Tideglusib is a non-competitive GSK-3β inhibitor which contain 1,2,4-thiadiazolidine-3,5-dione moiety, and now mainly used for progressive supranuclear palsy due to the lack of some primary cognitive endpoints and secondary endpoints in a phase IIb trail for Alzheimer's disease. Additionally, insufficient evidence exists to support that there are obvious covalent bonds between Tideglusib and GSK-3β. Targeted covalent inhibition strategy could improve the binding efficiency, selectivity and duration of kinase inhibitors. Based on the above premise, two series of targeted compounds with acryloyl warheads were designed and synthesized. The kinase inhibitory activity of the selected compound 10a with better neuroprotective effect improved 2.7 fold than that of Tideglusib. After the preliminary screening of GSK-3β inhibition and neuroprotective activity, the mechanism action of the selected compound 10a was investigated in vitro and in vivo. The results confirmed that 10a with excellent selectivity among the whole tested kinases could significantly reduce the expressions of APP and p-Tau via increasing the level of p-GSK-3β. The pharmacodynamic assay in vivo showed that 10a could markedly improve the learning and memory functions in AD mice induced by AlCl3 combined with d-galactose. At the same time, the damage of hippocampal neurons in AD mice was obviously reduced. Accordingly, the introduction of acryloyl warheads could increase the GSK-3β inhibitory activity of 1,2,4-thiadiazolidine-3,5-dione derivatives, and the selected compound 10a deserves further research as an effective GSK-3β inhibitor for the potential treatment of AD.
Collapse
Affiliation(s)
- Yongxi Dong
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China.
| | - Jun Lu
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China
| | - Shanhui Zhang
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China
| | - Lina Chen
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China
| | - Jinlan Wen
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China
| | - Fang Wang
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China
| | - Yongqing Mao
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China
| | - Lei Li
- Guizhou provincial Center for Disease Control and Prevention, Guiyang 550004, China
| | - Jiquan Zhang
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China
| | - Shanggao Liao
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China.
| | - Li Dong
- School of Pharmacy, Guizhou Medical University, Guian New District 550025, China.
| |
Collapse
|
30
|
Fiock KL, Betters RK, Hefti MM. Thioflavin S Staining and Amyloid Formation Are Unique to Mixed Tauopathies. J Histochem Cytochem 2023; 71:73-86. [PMID: 36861683 PMCID: PMC10071402 DOI: 10.1369/00221554231158428] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/30/2023] [Indexed: 03/03/2023] Open
Abstract
Tau phosphorylation, aggregation, and toxicity are the main drivers of neurodegeneration in multiple tauopathies, including Alzheimer's disease (AD) and frontotemporal lobar degeneration with tau. Although aggregation and amyloid formation are often assumed to be synonymous, the ability of tau aggregates in different diseases to form amyloids in vivo has not been systematically studied. We used the amyloid dye Thioflavin S to look at tau aggregates in mixed tauopathies such as AD and primary age-related tauopathy, as well as pure 3R or 4R tauopathies such as Pick's disease, progressive supranuclear palsy, and corticobasal degeneration. We found that aggregates of tau protein only form thioflavin-positive amyloids in mixed (3R/4R), but not pure (3R or 4R), tauopathies. Interestingly, neither astrocytic nor neuronal tau pathology was thioflavin-positive in pure tauopathies. As most current positron emission tomography tracers are based on thioflavin derivatives, this suggests that they may be more useful for differential diagnosis than the identification of a general tauopathy. Our findings also suggest that thioflavin staining may have utility as an alternative to traditional antibody staining for distinguishing between tau aggregates in patients with multiple pathologies and that the mechanisms for tau toxicity may differ between different tauopathies.
Collapse
Affiliation(s)
- Kimberly L. Fiock
- Department of Pathology, University of Iowa,
Iowa City, Iowa
- Experimental Pathology Graduate Program,
University of Iowa, Iowa City, Iowa
- Iowa Neuroscience Institute, Iowa City,
Iowa
| | - Ryan K. Betters
- Department of Pathology, University of Iowa,
Iowa City, Iowa
- Interdisciplinary Neuroscience Graduate
Program, University of Iowa, Iowa City, Iowa
- Iowa Neuroscience Institute, Iowa City,
Iowa
| | - Marco M. Hefti
- Department of Pathology, University of Iowa,
Iowa City, Iowa
- Experimental Pathology Graduate Program,
University of Iowa, Iowa City, Iowa
- Iowa Neuroscience Institute, Iowa City,
Iowa
| |
Collapse
|
31
|
Polanco JC, Akimov Y, Fernandes A, Briner A, Hand GR, van Roijen M, Balistreri G, Götz J. CRISPRi screening reveals regulators of tau pathology shared between exosomal and vesicle-free tau. Life Sci Alliance 2023; 6:6/1/e202201689. [PMID: 36316035 PMCID: PMC9622425 DOI: 10.26508/lsa.202201689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022] Open
Abstract
The aggregation of the microtubule-associated protein tau is a defining feature of Alzheimer's disease and other tauopathies. Tau pathology is believed to be driven by free tau aggregates and tau carried within exosome-like extracellular vesicles, both of which propagate trans-synaptically and induce tau pathology in recipient neurons by a corrupting process of seeding. Here, we performed a genome-wide CRISPRi screen in tau biosensor cells and identified cellular regulators shared by both mechanisms of tau seeding. We identified ANKLE2, BANF1, NUSAP1, EIF1AD, and VPS18 as the top validated regulators that restrict tau aggregation initiated by both exosomal and vesicle-free tau seeds. None of our validated hits affected the uptake of either form of tau seeds, supporting the notion that they operate through a cell-autonomous mechanism downstream of the seed uptake. Lastly, validation studies with human brain tissue also revealed that several of the identified protein hits are down-regulated in the brains of Alzheimer's patients, suggesting that their decreased activity may be required for the emergence or progression of tau pathology in the human brain.
Collapse
Affiliation(s)
- Juan Carlos Polanco
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Yevhen Akimov
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Avinash Fernandes
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Adam Briner
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Gabriel Rhys Hand
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | | | - Giuseppe Balistreri
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
32
|
Condello C, Merz GE, Aoyagi A, DeGrado WF, Prusiner SB. Aβ and Tau Prions Causing Alzheimer's Disease. Methods Mol Biol 2023; 2561:293-337. [PMID: 36399277 DOI: 10.1007/978-1-0716-2655-9_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Studies show that patients with Alzheimer's disease (AD) have both Aβ and tau prions, and thus, AD is a double-prion disease. AD patients with the greatest longevity exhibited low levels of both Aβ and tau prions; tau prions were nearly absent in the brains of almost half of the patients who lived beyond 80 years of age. Using cellular bioassays for prions in postmortem samples, we found that both Aβ and tau proteins misfold into prions leading to AD, which is either a sporadic or familial dementing disorder. Although AD is transmissible experimentally, there is no evidence that AD is either communicable or contagious. Since the progression of AD correlates poorly with insoluble Aβ in the central nervous system (CNS), it was difficult to distinguish between inert amyloids and Aβ prions. To measure the progression of AD, we devised rapid bioassays to measure the abundance of isoform-specific Aβ prions in the brains of transgenic (Tg) mice and in postmortem human CNS samples from AD victims and people who died of other neurodegenerative diseases (NDs). We found significant correlations between the longevity of individuals with AD, sex, and genetic background, despite the fact that all postmortem brain tissue had essentially the same confirmed neuropathology.Although brains from all AD patients had measurable levels of Aβ prions at death, the oldest individuals had lower Aβ prion levels than the younger ones. Additionally, the long-lived individuals had low tau prion levels that correlated with the extent of phosphorylated tau (p-tau). Unexpectedly, a longevity-dependent decrease in tau prions was found in spite of increasing amounts of total insoluble tau. When corrected for the abundance of insoluble tau, the tau prion levels decreased exponentially with respect to the age at death with a half-time of approximately one decade, and this correlated with the abundance of phosphorylated tau.Even though our findings with tau prions were not unexpected, they were counterintuitive; thus, tau phosphorylation and tau prion activity decreased exponentially with longevity in patients with AD ranging from ages 37 to 99 years. Our findings demonstrated an inverse correlation between longevity in AD patients and the abundance of neurotoxic tau prions. Moreover, our discovery may have profound implications for the selection of phenotypically distinct patient populations and the development of diagnostics and effective therapeutics for AD.
Collapse
Affiliation(s)
- Carlo Condello
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| | - Gregory E Merz
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Atsushi Aoyagi
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - William F DeGrado
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
33
|
Tzioras M, McGeachan RI, Durrant CS, Spires-Jones TL. Synaptic degeneration in Alzheimer disease. Nat Rev Neurol 2023; 19:19-38. [PMID: 36513730 DOI: 10.1038/s41582-022-00749-z] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 12/15/2022]
Abstract
Alzheimer disease (AD) is characterized by progressive cognitive decline in older individuals accompanied by the presence of two pathological protein aggregates - amyloid-β and phosphorylated tau - in the brain. The disease results in brain atrophy caused by neuronal loss and synapse degeneration. Synaptic loss strongly correlates with cognitive decline in both humans and animal models of AD. Indeed, evidence suggests that soluble forms of amyloid-β and tau can cause synaptotoxicity and spread through neural circuits. These pathological changes are accompanied by an altered phenotype in the glial cells of the brain - one hypothesis is that glia excessively ingest synapses and modulate the trans-synaptic spread of pathology. To date, effective therapies for the treatment or prevention of AD are lacking, but understanding how synaptic degeneration occurs will be essential for the development of new interventions. Here, we highlight the mechanisms through which synapses degenerate in the AD brain, and discuss key questions that still need to be answered. We also cover the ways in which our understanding of the mechanisms of synaptic degeneration is leading to new therapeutic approaches for AD.
Collapse
Affiliation(s)
- Makis Tzioras
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Robert I McGeachan
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.,The Hospital for Small Animals, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - Claire S Durrant
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK. .,UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
34
|
Bittar A, Al-Lahham R, Bhatt N, Moore K, Montalbano M, Jerez C, Fung L, McAllen S, Ellsworth A, Kayed R. Passive Immunotherapy Targeting Tau Oligomeric Strains Reverses Tauopathy Phenotypes in Aged Human-Tau Mice in a Mouse Model-Specific Manner. J Alzheimers Dis 2022; 90:1103-1122. [DOI: 10.3233/jad-220518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Tau oligomers are one of the most toxic species, displaying prion-like strains which have different conformations resulting in different tauopathies. Passive immunotherapy targeting different tau species is a promising therapeutic approach. Age is one of the greatest risk factors; however, most immunotherapy studies are done in young to middle-aged mice tauopathy models, which is not representative of the many clinical trials done with older humans with established tauopathies. Objective: We utilized two different clones of tau oligomer monoclonal antibodies (TOMAs) in aged Htau and JNPL3 mouse models to investigate the potential of passive immunotherapy. Methods: Aged mice received a single intravenous injection of 120 μg/animal of either TOMA1, TOMA3 clones or a non-specific IgG. Their cognitive functions were assessed one-week post-injection using Y-maze and novel object recognition tests. Brain tissues were analyzed using biochemical and immunological assays. Results: TOMA 1 and 3 rescues cognitive phenotypes in aged animals in a mouse model-specific manner, indicative by a reduction in tau oligomers levels. The TOMAs were shown to have strong reactivity with different tau oligomeric species in the different mouse models in vitro and ex vivo. Conclusion: This is the first study testing tau passive immunotherapy in aged animals and supports our previous reports on of the role of oligomeric tau in disease progression further validating the potential of TOMAs to rescue the late-stage disease pathology and phenotype. Moreover, this study suggests that multiple tau oligomeric strains exist in aged animals; therefore, it is of great importance to further characterize these strains.
Collapse
Affiliation(s)
- Alice Bittar
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rabab Al-Lahham
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kenya Moore
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Leiana Fung
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Anna Ellsworth
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
35
|
Hromadkova L, Siddiqi MK, Liu H, Safar JG. Populations of Tau Conformers Drive Prion-like Strain Effects in Alzheimer's Disease and Related Dementias. Cells 2022; 11:2997. [PMID: 36230957 PMCID: PMC9562632 DOI: 10.3390/cells11192997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Recent findings of diverse populations of prion-like conformers of misfolded tau protein expand the prion concept to Alzheimer's disease (AD) and monogenic frontotemporal lobar degeneration (FTLD)-MAPT P301L, and suggest that distinct strains of misfolded proteins drive the phenotypes and progression rates in many neurodegenerative diseases. Notable progress in the previous decades has generated many lines of proof arguing that yeast, fungal, and mammalian prions determine heritable as well as infectious traits. The extraordinary phenotypic diversity of human prion diseases arises from structurally distinct prion strains that target, at different progression speeds, variable brain structures and cells. Although human prion research presents beneficial lessons and methods to study the mechanism of strain diversity of protein-only pathogens, the fundamental molecular mechanism by which tau conformers are formed and replicate in diverse tauopathies is still poorly understood. In this review, we summarize up to date advances in identification of diverse tau conformers through biophysical and cellular experimental paradigms, and the impact of heterogeneity of pathological tau strains on personalized structure- and strain-specific therapeutic approaches in major tauopathies.
Collapse
Affiliation(s)
- Lenka Hromadkova
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - He Liu
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jiri G. Safar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
36
|
Odfalk KF, Bieniek KF, Hopp SC. Microglia: Friend and foe in tauopathy. Prog Neurobiol 2022; 216:102306. [PMID: 35714860 PMCID: PMC9378545 DOI: 10.1016/j.pneurobio.2022.102306] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/24/2022] [Accepted: 06/10/2022] [Indexed: 12/16/2022]
Abstract
Aggregation of misfolded microtubule associated protein tau into abnormal intracellular inclusions defines a class of neurodegenerative diseases known as tauopathies. The consistent spatiotemporal progression of tau pathology in Alzheimer's disease (AD) led to the hypothesis that tau aggregates spread in the brain via bioactive tau "seeds" underlying advancing disease course. Recent studies implicate microglia, the resident immune cells of the central nervous system, in both negative and positive regulation of tau pathology. Polymorphisms in genes that alter microglial function are associated with the development of AD and other tauopathies. Experimental manipulation of microglia function can alter tau pathology and microglia-mediated neuroinflammatory cascades can exacerbate tau pathology. Microglia also exert protective functions by mitigating tau spread: microglia internalize tau seeds and have the capacity to degrade them. However, when microglia fail to degrade these tau seeds there are deleterious consequences, including secretion of exosomes containing tau that can spread to neurons. This review explores the intersection of microglia and tau from the perspective of neuropathology, neuroimaging, genetics, transcriptomics, and molecular biology. As tau-targeted therapies such as anti-tau antibodies advance through clinical trials, it is critical to understand the interaction between tau and microglia.
Collapse
Affiliation(s)
- Kristian F Odfalk
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
37
|
Zeng Z, Fichou Y, Vigers M, Tsay K, Han S. Illuminating the Structural Basis of Tau Aggregation by Intramolecular Distance Tracking: A Perspective on Methods. J Phys Chem B 2022; 126:6384-6395. [PMID: 35994024 DOI: 10.1021/acs.jpcb.2c02022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The aggregation of the tau protein is central to several neurodegenerative diseases, collectively known as tauopathies. High-resolution views of tau tangles accumulated under pathological conditions in post-mortem brains have been revealed recently by cryogenic electron microscopy. One of the striking discoveries was that fibril folds are unique to and homogeneous within one disease family, but typically different between different tauopathies. It is widely believed that seeded aggregation can achieve structural propagation of tau fibrils and generate pathological fibril structures. However, direct molecular level measurement of structural evolution during aggregation is missing. Here, we discuss our perspective on the biophysical approaches that can contribute to the ongoing debate regarding the prion-like propagation of tau and the role of cofactors. We discuss the unique potential of double electron-electron resonance (DEER)-based intramolecular distance measurement, sensitive to two to several nanometers distances. DEER can track the structural evolution of tau along the course of aggregation from the completely disordered state, to partially ordered and highly ordered fibril states, and has the potential to be a key tool to elucidate the disease-specific tau aggregation pathways.
Collapse
Affiliation(s)
- Zhikai Zeng
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Yann Fichou
- CNRS, Chemistry and Biology of Membranes and Nanoobjects (CBMN) UMR 5348, Institut Europeen de Chimie et Biologie (IECB), University of Bordeaux, 33600 Pessac, France
| | - Michael Vigers
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Karen Tsay
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Songi Han
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States.,Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
38
|
Huynh MB, Rebergue N, Merrick H, Gomez-Henao W, Jospin E, Biard DSF, Papy-Garcia D. HS3ST2 expression induces the cell autonomous aggregation of tau. Sci Rep 2022; 12:10850. [PMID: 35760982 PMCID: PMC9237029 DOI: 10.1038/s41598-022-13486-6] [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: 05/30/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
Heparan sulfates have long been known to intracellularly accumulate in Alzheimer's disease neurons, where they colocalize with neurofibrillary tangles made of abnormally phosphorylated and aggregated tau protein. However, the reasons and consequences of the heparan sulfates accumulation in the Alzheimer's cells are not yet well understood. Previously, we showed that the neural heparan sulfate 3-O-sulfotransferase HS3ST2 is critical for the abnormal phosphorylation of tau in Alzheimer's disease-related tauopathy. Using cell models of tauopathy we showed that intracellular 3-O-sulfatated heparan sulfates interact with tau inducing its abnormal phosphorylation. However, it is unknown whether HS3ST2 expression induces the intracellular aggregation of tau in cells. Here, by using replicative pEBV plasmids, we engineered HEK293 cells to stably express HS3ST2 together with human tau carrying or not the P301S mutation. We show that HS3ST2 gain of function induces the cell autonomous aggregation of tau not only in cells expressing tauP301S, but also in cells expressing the wild type tau. Our engineered cells mimicked both the HS intracellular accumulation observed in neurons of Alzheimer's disease and the tau aggregation characteristic of tauopathy development and evolution. These results give evidence that the neural HS3ST2 plays a critical role in the cell autonomous self-aggregation of tau.
Collapse
Affiliation(s)
- M B Huynh
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Univ Paris Est Creteil (UPEC), F-94010 Creteil, France
| | - N Rebergue
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Univ Paris Est Creteil (UPEC), F-94010 Creteil, France
| | - H Merrick
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Univ Paris Est Creteil (UPEC), F-94010 Creteil, France
| | - W Gomez-Henao
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Univ Paris Est Creteil (UPEC), F-94010 Creteil, France
- Departamento de Bioquímica, Laboratorio Internacional Gly-CRRET-UNAM, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - E Jospin
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Univ Paris Est Creteil (UPEC), F-94010 Creteil, France
| | - D S F Biard
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Univ Paris Est Creteil (UPEC), F-94010 Creteil, France
- CEA, Institut de Biologie François Jacob (IBFJ), SEPIA, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - D Papy-Garcia
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Univ Paris Est Creteil (UPEC), F-94010 Creteil, France.
| |
Collapse
|
39
|
Anti-fibrillization Effects of Sulfonamide Derivatives on α-Synuclein and Hyperphosphorylated Tau Isoform 1N4R. J Mol Struct 2022; 1267. [DOI: 10.1016/j.molstruc.2022.133574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
40
|
Dregni AJ, Duan P, Xu H, Changolkar L, El Mammeri N, Lee VMY, Hong M. Fluent molecular mixing of Tau isoforms in Alzheimer's disease neurofibrillary tangles. Nat Commun 2022; 13:2967. [PMID: 35624093 PMCID: PMC9142584 DOI: 10.1038/s41467-022-30585-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/04/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is defined by intracellular neurofibrillary tangles formed by the microtubule-associated protein tau and extracellular plaques formed by the β-amyloid peptide. AD tau tangles contain a mixture of tau isoforms with either four (4R) or three (3R) microtubule-binding repeats. Here we use solid-state NMR to determine how 4R and 3R tau isoforms mix at the molecular level in AD tau aggregates. By seeding differentially isotopically labeled 4R and 3R tau monomers with AD brain-derived tau, we measured intermolecular contacts of the two isoforms. The NMR data indicate that 4R and 3R tau are well mixed in the AD-tau seeded fibrils, with a 60:40 incorporation ratio of 4R to 3R tau and a small homotypic preference. The AD-tau templated 4R tau, 3R tau, and mixed 4R and 3R tau fibrils exhibit no structural differences in the rigid β-sheet core or the mobile domains. Therefore, 4R and 3R tau are fluently recruited into the pathological fold of AD tau aggregates, which may explain the predominance of AD among neurodegenerative disorders.
Collapse
Affiliation(s)
- Aurelio J Dregni
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA
| | - Pu Duan
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA
| | - Hong Xu
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Lakshmi Changolkar
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Nadia El Mammeri
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
41
|
Just MK, Gram H, Theologidis V, Jensen PH, Nilsson KPR, Lindgren M, Knudsen K, Borghammer P, Van Den Berge N. Alpha-Synuclein Strain Variability in Body-First and Brain-First Synucleinopathies. Front Aging Neurosci 2022; 14:907293. [PMID: 35693346 PMCID: PMC9178288 DOI: 10.3389/fnagi.2022.907293] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Pathogenic alpha-synuclein (asyn) aggregates are a defining feature of neurodegenerative synucleinopathies, which include Parkinson's disease, Lewy body dementia, pure autonomic failure and multiple system atrophy. Early accurate differentiation between these synucleinopathies is challenging due to the highly heterogeneous clinical profile at early prodromal disease stages. Therefore, diagnosis is often made in late disease stages when a patient presents with a broad range of motor and non-motor symptoms easing the differentiation. Increasing data suggest the clinical heterogeneity seen in patients is explained by the presence of distinct asyn strains, which exhibit variable morphologies and pathological functions. Recently, asyn seed amplification assays (PMCA and RT-QuIC) and conformation-specific ligand assays have made promising progress in differentiating between synucleinopathies in prodromal and advanced disease stages. Importantly, the cellular environment is known to impact strain morphology. And, asyn aggregate pathology can propagate trans-synaptically along the brain-body axis, affecting multiple organs and propagating through multiple cell types. Here, we present our hypothesis that the changing cellular environments, an asyn seed may encounter during its brain-to-body or body-to-brain propagation, may influence the structure and thereby the function of the aggregate strains developing within the different cells. Additionally, we aim to review strain characteristics of the different synucleinopathies in clinical and preclinical studies. Future preclinical animal models of synucleinopathies should investigate if asyn strain morphology is altered during brain-to-body and body-to-brain spreading using these seeding amplification and conformation-specific assays. Such findings would greatly deepen our understanding of synucleinopathies and the potential link between strain and phenotypic variability, which may enable specific diagnosis of different synucleinopathies in the prodromal phase, creating a large therapeutic window with potential future applications in clinical trials and personalized therapeutics.
Collapse
Affiliation(s)
- Mie Kristine Just
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Hjalte Gram
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Vasileios Theologidis
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Poul Henning Jensen
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - K. Peter R. Nilsson
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Mikael Lindgren
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karoline Knudsen
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Per Borghammer
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Nathalie Van Den Berge
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
42
|
Annadurai N, Malina L, Malohlava J, Hajdúch M, Das V. Tau R2 and R3 are essential regions for tau aggregation, seeding and propagation. Biochimie 2022; 200:79-86. [PMID: 35623497 DOI: 10.1016/j.biochi.2022.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022]
Abstract
Tauopathies are characterised by intracellular deposits of fibrillar tau tangles. However, the interneuronal spread of pathological tau species precedes the development of major tau burdens. Two amyloid motifs, VQIINK in repeat 2 and VQIVYK in repeat 3, of tau repeat domain, assemble into β-sheet-rich fibrils on their own but alone do not form seed-competent fibrils. In contrast, the entire R3 region self-aggregates and forms seed-competent fibrils. Our study aimed to identify the minimal regions in the tau repeat domain that define seeding and its impact on intracellular tau phosphorylation and aggregation. Using peptides of individual repeats, we show that R2, like R3, forms seed-competent fibrils when assembled in the presence of heparin. However, R3, but not R2, forms seed-competent fibrils when assembled without heparin, even though both R2 and R3 have identical N-terminal hexapeptide and cysteine residue sequences. Moreover, cysteine to alanine substitution in R3 abrogates its self-aggregation and seeding potency. Tau RD P301S biosensor cells and Tau P301L (0N4R)-expressing HEK293 cells seeded with R2 and R3 fibrils show the induction of pathological phosphorylation of tau at Ser262/Ser396/Ser404 positions and oligomerisation of native tau. Protein fractions of biosensor cells seeded with R2 and R3 fibrils reseed endogenous tau aggregation when introduced into a fresh set of biosensor cells. Our findings suggest that R3 may be the minimal region for pathological seed generation under physiological conditions, whereas R2 might need polyanionic cofactors to generate pathogenic seeds. Lastly, R2 and R3 fibrils induce template-induced misfolding and pathological hyperphosphorylation of intracellular tau, making intracellular tau seed-competent.
Collapse
Affiliation(s)
- Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 77900, Olomouc, Czech Republic
| | - Lukáš Malina
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacký University in Olomouc, Olomouc, Czech Republic
| | - Jakub Malohlava
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacký University in Olomouc, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 77900, Olomouc, Czech Republic; Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Krizkovskeho 511/8, 77900, Olomouc, Czech Republic
| | - Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 1333/5, 77900, Olomouc, Czech Republic; Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Krizkovskeho 511/8, 77900, Olomouc, Czech Republic.
| |
Collapse
|
43
|
Han ZZ, Kang SG, Arce L, Westaway D. Prion-like strain effects in tauopathies. Cell Tissue Res 2022; 392:179-199. [PMID: 35460367 PMCID: PMC9034081 DOI: 10.1007/s00441-022-03620-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
Tau is a microtubule-associated protein that plays crucial roles in physiology and pathophysiology. In the realm of dementia, tau protein misfolding is associated with a wide spectrum of clinicopathologically diverse neurodegenerative diseases, collectively known as tauopathies. As proposed by the tau strain hypothesis, the intrinsic heterogeneity of tauopathies may be explained by the existence of structurally distinct tau conformers, “strains”. Tau strains can differ in their associated clinical features, neuropathological profiles, and biochemical signatures. Although prior research into infectious prion proteins offers valuable lessons for studying how a protein-only pathogen can encompass strain diversity, the underlying mechanism by which tau subtypes are generated remains poorly understood. Here we summarize recent advances in understanding different tau conformers through in vivo and in vitro experimental paradigms, and the implications of heterogeneity of pathological tau species for drug development.
Collapse
Affiliation(s)
- Zhuang Zhuang Han
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Luis Arce
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada. .,Department of Medicine, University of Alberta, Edmonton, AB, Canada. .,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
44
|
Martin W, Sheynkman G, Lightstone FC, Nussinov R, Cheng F. Interpretable artificial intelligence and exascale molecular dynamics simulations to reveal kinetics: Applications to Alzheimer's disease. Curr Opin Struct Biol 2022; 72:103-113. [PMID: 34628220 PMCID: PMC8860862 DOI: 10.1016/j.sbi.2021.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 02/03/2023]
Abstract
The rapid increase in computing power, especially with the integration of graphics processing units, has dramatically increased the capabilities of molecular dynamics simulations. To date, these capabilities extend from running very long simulations (tens to hundreds of microseconds) to thousands of short simulations. However, the expansive data generated in these simulations must be made interpretable not only by the investigator who performs them but also by others as well. Here, we demonstrate how integrating learning techniques, such as artificial intelligence, machine learning, and neural networks, into analysis pipelines can reveal the kinetics of Alzheimer's disease (AD) protein aggregation. We review select AD targets, describe current simulation methods, and introduce learning concepts and their application in AD, highlighting limitations and potential solutions.
Collapse
Affiliation(s)
- William Martin
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Gloria Sheynkman
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Felice C Lightstone
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Lab, Livermore, CA, 94550, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
45
|
Roda AR, Serra-Mir G, Montoliu-Gaya L, Tiessler L, Villegas S. Amyloid-beta peptide and tau protein crosstalk in Alzheimer's disease. Neural Regen Res 2022; 17:1666-1674. [PMID: 35017413 PMCID: PMC8820696 DOI: 10.4103/1673-5374.332127] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease is a neurodegenerative disease that accounts for most of the 50-million dementia cases worldwide in 2018. A large amount of evidence supports the amyloid cascade hypothesis, which states that amyloid-beta accumulation triggers tau hyperphosphorylation and aggregation in form of neurofibrillary tangles, and these aggregates lead to inflammation, synaptic impairment, neuronal loss, and thus to cognitive decline and behavioral abnormalities. The poor correlation found between cognitive decline and amyloid plaques, have led the scientific community to question whether amyloid-beta accumulation is actually triggering neurodegeneration in Alzheimer’s disease. The occurrence of tau neurofibrillary tangles better correlates to neuronal loss and clinical symptoms and, although amyloid-beta may initiate the cascade of events, tau impairment is likely the effector molecule of neurodegeneration. Recently, it has been shown that amyloid-beta and tau cooperatively work to impair transcription of genes involved in synaptic function and, more importantly, that downregulation of tau partially reverses transcriptional perturbations. Despite mounting evidence points to an interplay between amyloid-beta and tau, some factors could independently affect both pathologies. Thus, the dual pathway hypothesis, which states that there are common upstream triggers causing both amyloid-beta and tau abnormalities has been proposed. Among others, the immune system seems to be strongly involved in amyloid-beta and tau pathologies. Other factors, as the apolipoprotein E ε4 isoform has been suggested to act as a link between amyloid-beta and tau hyperphosphorylation. Interestingly, amyloid-beta-immunotherapy reduces not only amyloid-beta but also tau levels in animal models and in clinical trials. Likewise, it has been shown that tau-immunotherapy also reduces amyloid-beta levels. Thus, even though amyloid-beta immunotherapy is more advanced than tau-immunotherapy, combined amyloid-beta and tau-directed therapies at early stages of the disease have recently been proposed as a strategy to stop the progression of Alzheimer’s disease.
Collapse
Affiliation(s)
- Alejandro R Roda
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Gabriel Serra-Mir
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Lidia Tiessler
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sandra Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
46
|
Fernandes AR, Dujardin S, Maté de Gérando A, Hyman BT, Frosch MP. Impact of Sterilization Methods on the Seeding Ability of Human Tau Proteopathic Seeds. J Neuropathol Exp Neurol 2021; 80:912-921. [PMID: 34498073 DOI: 10.1093/jnen/nlab087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The protein tau, when misfolded in neurodegenerative diseases, has several prion-like properties including being able to spread by cell-to-cell transfer, induce templated seeding, and exist in distinct conformational strains. These properties of transmission may present health hazards when lesion-containing biospecimens are used in research and neuropathology laboratories. We evaluated the impact standard sterilization and cleaning methods have on the capacity of tau seeds to induce aggregation. We employed a previously developed, highly sensitive FRET-based biosensor assay to assess remnant tau seeding after exposure to these procedures. For tau species derived from human Alzheimer disease tissue (brain homogenate and sarkosyl-insoluble fibrils), both autoclaving and incubation in 90.6% formic acid were sufficient to reduce tau bioactivity. By contrast, boiling was not always effective in completely blocking bioactivity in the seeding assay. Notably, only formic acid incubation was able to produce a similar reduction in tissue from a P301L mutant tau mouse model of tauopathy. Our study highlights nuances in methods for inactivation of tau seeding which may support adapted tissue processing procedures, especially in research settings. These findings also highlight the importance of universal precautions when handling human neuropathological and research laboratory materials.
Collapse
Affiliation(s)
- Analiese R Fernandes
- From the Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Simon Dujardin
- From the Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA (SD, AMdG, BTH, MPF)
| | - Anastasie Maté de Gérando
- From the Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA (SD, AMdG, BTH, MPF)
| | - Bradley T Hyman
- From the Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA (SD, AMdG, BTH, MPF).,C.S. Kubik Laboratory for Neuropathology, Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew P Frosch
- From the Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA (SD, AMdG, BTH, MPF).,C.S. Kubik Laboratory for Neuropathology, Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Sexton C, Snyder H, Beher D, Boxer AL, Brannelly P, Brion JP, Buée L, Cacace AM, Chételat G, Citron M, DeVos SL, Diaz K, Feldman HH, Frost B, Goate AM, Gold M, Hyman B, Johnson K, Karch CM, Kerwin DR, Koroshetz WJ, Litvan I, Morris HR, Mummery CJ, Mutamba J, Patterson MC, Quiroz YT, Rabinovici GD, Rommel A, Shulman MB, Toledo-Sherman LM, Weninger S, Wildsmith KR, Worley SL, Carrillo MC. Current directions in tau research: Highlights from Tau 2020. Alzheimers Dement 2021; 18:988-1007. [PMID: 34581500 DOI: 10.1002/alz.12452] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 11/07/2022]
Abstract
Studies supporting a strong association between tau deposition and neuronal loss, neurodegeneration, and cognitive decline have heightened the allure of tau and tau-related mechanisms as therapeutic targets. In February 2020, leading tau experts from around the world convened for the first-ever Tau2020 Global Conference in Washington, DC, co-organized and cosponsored by the Rainwater Charitable Foundation, the Alzheimer's Association, and CurePSP. Representing academia, industry, government, and the philanthropic sector, presenters and attendees discussed recent advances and current directions in tau research. The meeting provided a unique opportunity to move tau research forward by fostering global partnerships among academia, industry, and other stakeholders and by providing support for new drug discovery programs, groundbreaking research, and emerging tau researchers. The meeting also provided an opportunity for experts to present critical research-advancing tools and insights that are now rapidly accelerating the pace of tau research.
Collapse
Affiliation(s)
| | | | | | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Pat Brannelly
- Alzheimer's Disease Data Initiative, Kirkland, WI, USA
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Luc Buée
- Univ Lille, Inserm, CHU-Lille, Lille Neuroscience and Cognition, Place de Verdun, Lille, France
| | | | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, France
| | - Martin Citron
- Neuroscience TA, Braine l'Alleud, UCB Biopharma, Brussels, Belgium
| | - Sarah L DeVos
- Translational Sciences, Denali Therapeutics, San Francisco, California, USA
| | | | - Howard H Feldman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Bess Frost
- Sam & Ann Barshop Institute for Longevity and Aging Studies, Glenn Biggs Institute for Alzheimer's & Neurodegenerative Disorders, Department of Cell Systems & Anatomy, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Neuroscience, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Gold
- AbbVie, Neurosciences Development, North Chicago, Illinois, USA
| | - Bradley Hyman
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Johnson
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Diana R Kerwin
- Kerwin Medical Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Walter J Koroshetz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, San Diego, California, USA
| | - Huw R Morris
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | - Catherine J Mummery
- Dementia Research Centre, National Hospital for Neurology and Neurosurgery, University College London, London, UK
| | | | - Marc C Patterson
- Departments of Neurology, Pediatrics and Medical Genetics, Mayo Clinic, Rochester, Minnesota, USA
| | - Yakeel T Quiroz
- Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Departments of Neurology, Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Amy Rommel
- Tau Consortium, Rainwater Charitable Foundation, Fort Worth, Texas, USA
| | - Melanie B Shulman
- Neurodegeneration Development Unit, Biogen, Boston, Massachusetts, USA
| | | | | | - Kristin R Wildsmith
- Department of Biomarker Development, Genentech, South San Francisco, California, USA
| | - Susan L Worley
- Independent science writer, Bryn Mawr, Pennsylvania, USA
| | | |
Collapse
|
48
|
Kroth H, Oden F, Molette J, Schieferstein H, Gabellieri E, Mueller A, Berndt M, Sreenivasachary N, Serra AM, Capotosti F, Schmitt-Willich H, Hickman D, Pfeifer A, Dinkelborg L, Stephens A. PI-2620 Lead Optimization Highlights the Importance of Off-Target Assays to Develop a PET Tracer for the Detection of Pathological Aggregated Tau in Alzheimer's Disease and Other Tauopathies. J Med Chem 2021; 64:12808-12830. [PMID: 34455780 DOI: 10.1021/acs.jmedchem.1c00861] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The first candidate PI-2014 was tested in healthy controls and subjects with Alzheimer's disease (AD). As PI-2014 displayed off-target binding to monoamine oxidase A (MAO-A), a new lead with improved binding to Tau and decreased MAO-A binding was required. For compound optimization, Tau binding assays based on both human AD brain homogenate and Tau-paired helical filaments were employed. Furthermore, two MAO-A screening assays based on (1) human-recombinant MAO-A and (2) displacement of 2-fluoro-ethyl-harmine from mouse brain homogenate were employed. Removing the N-methyl group from the tricyclic core resulted in compounds displaying improved Tau binding. For the final round of optimization, the cyclic amine substituents were replaced by pyridine derivatives. PI-2620 (2-(2-fluoropyridin-4-yl)-9H-pyrrolo[2,3-b:4,5-c']dipyridine) emerged as a best candidate displaying high Tau binding, low MAO-A binding, high brain uptake, and fast and complete brain washout. Furthermore, PI-2620 showed Tau binding on brain sections from corticobasal degeneration, progressive supranuclear palsy, and Pick's disease.
Collapse
Affiliation(s)
- Heiko Kroth
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Felix Oden
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Jerome Molette
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | | | | | - Andre Mueller
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Mathias Berndt
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | | | | | | | | | - David Hickman
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Andrea Pfeifer
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Ludger Dinkelborg
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Andrew Stephens
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| |
Collapse
|
49
|
DnaJC7 binds natively folded structural elements in tau to inhibit amyloid formation. Nat Commun 2021; 12:5338. [PMID: 34504072 PMCID: PMC8429438 DOI: 10.1038/s41467-021-25635-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
Molecular chaperones, including Hsp70/J-domain protein (JDP) families, play central roles in binding substrates to prevent their aggregation. How JDPs select different conformations of substrates remains poorly understood. Here, we report an interaction between the JDP DnaJC7 and tau that efficiently suppresses tau aggregation in vitro and in cells. DnaJC7 binds preferentially to natively folded wild-type tau, but disease-associated mutants in tau reduce chaperone binding affinity. We identify that DnaJC7 uses a single TPR domain to recognize a β-turn structural element in tau that contains the 275VQIINK280 amyloid motif. Wild-type tau, but not mutant, β-turn structural elements can block full-length tau binding to DnaJC7. These data suggest DnaJC7 preferentially binds and stabilizes natively folded conformations of tau to prevent tau conversion into amyloids. Our work identifies a novel mechanism of tau aggregation regulation that can be exploited as both a diagnostic and a therapeutic intervention.
Collapse
|
50
|
Wang D, Huang X, Yan L, Zhou L, Yan C, Wu J, Su Z, Huang Y. The Structure Biology of Tau and Clue for Aggregation Inhibitor Design. Protein J 2021; 40:656-668. [PMID: 34401998 DOI: 10.1007/s10930-021-10017-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
Tau is a microtubule-associated protein that is mainly expressed in central and peripheral nerve systems. Tau binds to tubulin and regulates assembly and stabilization of microtubule, thus playing a critical role in neuron morphology, axon development and navigation. Tau is highly stable under normal conditions; however, there are several factors that can induce or promote aggregation of tau, forming neurofibrillary tangles. Neurofibrillary tangles are toxic to neurons, which may be related to a series of neurodegenerative diseases including Alzheimer's disease. Thus, tau is widely accepted as an important therapeutic target for neurodegenerative diseases. While the monomeric structure of tau is highly disordered, the aggregate structure of tau is formed by closed packing of β-stands. Studies on the structure of tau and the structural transition mechanism provide valuable information on the occurrence, development, and therapy of tauopathies. In this review, we summarize recent progress on the structural investigation of tau and based on which we discuss aggregation inhibitor design.
Collapse
Affiliation(s)
- Dan Wang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Xianlong Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Lu Yan
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Luoqi Zhou
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Chang Yan
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Jinhu Wu
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Zhengding Su
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Yongqi Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China. .,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| |
Collapse
|