1
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Singh R, Panghal A, Jadhav K, Thakur A, Verma RK, Singh C, Goyal M, Kumar J, Namdeo AG. Recent Advances in Targeting Transition Metals (Copper, Iron, and Zinc) in Alzheimer's Disease. Mol Neurobiol 2024; 61:10916-10940. [PMID: 38809370 DOI: 10.1007/s12035-024-04256-8] [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: 01/05/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Changes in the transition metal homeostasis in the brain are closely linked with Alzheimer's disease (AD), including intraneuronal iron accumulation and extracellular copper and zinc pooling in the amyloid plague. The brain copper, zinc, and iron surplus are commonly acknowledged characteristics of AD, despite disagreements among some. This has led to the theory that oxidative stress resulting from abnormal homeostasis of these transition metals may be a causative explanation behind AD. In the nervous system, the interaction of metals with proteins appears to be an essential variable in the development or suppression of neurodegeneration. Chelation treatment may be an option for treating neurodegeneration induced by transition metal ion dyshomeostasis. Some clinicians even recommend using chelating agents as an adjunct therapy for AD. The current review also looks at the therapeutic strategies that have been attempted, primarily with metal-chelating drugs. Metal buildup in the nervous system, as reported in the AD, could be the result of compensatory mechanisms designed to improve metal availability for physiological functions.
Collapse
Affiliation(s)
- Raghuraj Singh
- Pharmaceutical Nanotechnology Lab, Institutes of Nano Science and Technology (INST), Sector 81. Mohali, Punjab, 140306, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Archna Panghal
- Department of Pharmacology and Toxicology, Facility for Risk Assessment and Intervention Studies, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, India
| | - Krishna Jadhav
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Ashima Thakur
- Faculty of Pharmaceutical Sciences, ICFAI University, Baddi, Distt. Solan, Himachal Pradesh, 174103, India
| | - Rahul Kumar Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Charan Singh
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| | - Manoj Goyal
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| | - Jayant Kumar
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India.
| | - Ajay G Namdeo
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| |
Collapse
|
3
|
Кондаурова ЕМ, Komarova AA, Ilchibaeva TV, Rodnyy AY, Zalivina EA, Naumenko VS. Effect of amisulpride on the expression of serotonin receptors, neurotrophic factor BDNF and its receptors in mice with overexpression of the aggregation-prone [R406W] mutant tau protein. Vavilovskii Zhurnal Genet Selektsii 2024; 28:398-406. [PMID: 39027123 PMCID: PMC11253013 DOI: 10.18699/vjgb-24-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 07/20/2024] Open
Abstract
Serotonin 5-HT7 receptors (5-HT7R) are attracting increasing attention as important participants in the mechanisms of Alzheimer's disease and as a possible target for the treatment of various tau pathologies. In this study, we investigated the effects of amisulpride (5-HT7R inverse agonist) in C57BL/6J mice with experimentally induced expression of the gene encoding the aggregation-prone human Tau[R406W] protein in the prefrontal cortex. In these animals we examined short-term memory and the expression of genes involved in the development of tauopathy (Htr7 and Cdk5), as well as biomarkers of neurodegenerative processes - the Bdnf gene and its receptors TrkB (the Ntrk2 gene) and p75NTR (the Ngfr gene). In a short-term memory test, there was no difference in the discrimination index between mice treated with AAV-Tau[R406W] and mice treated with AAV-EGFP. Amisulpride did not affect this parameter. Administration of AAV-Tau[R406W] resulted in increased expression of the Htr7, Htr1a, and Cdk5 genes in the prefrontal cortex compared to AAV-EGFP animals. At the same time, amisulpride at the dose of 10 mg/kg in animals from the AAV-Tau[R406W] group caused a decrease in the Htr7, Htr1a genes mRNA levels compared to animals from the AAV-Tau[R406W] group treated with saline. A decrease in the expression of the Bdnf and Ntrk2 genes in the prefrontal cortex was revealed after administration of AAV-Tau[R406W]. Moreover, amisulpride at various doses (3 and 10 mg/kg) caused the same decrease in the transcription of these genes in mice without tauopathy. It is also interesting that in mice of the AAV-EGFP group, administration of amisulpride at the dose of 10 mg/kg increased the Ngfr gene mRNA level. The data obtained allow us to propose the use of amisulpride in restoring normal tau protein function. However, it should be noted that prolonged administration may result in adverse effects such as an increase in Ngfr expression and a decrease in Bdnf and Ntrk2 expression, which is probably indicative of an increase in neurodegenerative processes.
Collapse
Affiliation(s)
- Е М Кондаурова
- Федеральный исследовательский центр Институт цитологии и генетики Сибирского отделения Российской академии наук, Новосибирск, Россия
| | - A A Komarova
- Федеральный исследовательский центр Институт цитологии и генетики Сибирского отделения Российской академии наук, Новосибирск, Россия
| | - T V Ilchibaeva
- Федеральный исследовательский центр Институт цитологии и генетики Сибирского отделения Российской академии наук, Новосибирск, Россия
| | - A Ya Rodnyy
- Федеральный исследовательский центр Институт цитологии и генетики Сибирского отделения Российской академии наук, Новосибирск, Россия
| | - E A Zalivina
- Федеральный исследовательский центр Институт цитологии и генетики Сибирского отделения Российской академии наук, Новосибирск, Россия
| | - V S Naumenko
- Федеральный исследовательский центр Институт цитологии и генетики Сибирского отделения Российской академии наук, Новосибирск, Россия
| |
Collapse
|
4
|
Pradeepkiran JA, Baig J, Islam MA, Kshirsagar S, Reddy PH. Amyloid-β and Phosphorylated Tau are the Key Biomarkers and Predictors of Alzheimer's Disease. Aging Dis 2024:AD.2024.0286. [PMID: 38739937 DOI: 10.14336/ad.2024.0286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
Alzheimer's disease (AD) is a age-related neurodegenerative disease and is a major public health concern both in Texas, US and Worldwide. This neurodegenerative disease is mainly characterized by amyloid-beta (Aβ) and phosphorylated Tau (p-Tau) accumulation in the brains of patients with AD and increasing evidence suggests that these are key biomarkers in AD. Both Aβ and p-tau can be detected through various imaging techniques (such as positron emission tomography, PET) and cerebrospinal fluid (CSF) analysis. The presence of these biomarkers in individuals, who are asymptomatic or have mild cognitive impairment can indicate an increased risk of developing AD in the future. Furthermore, the combination of Aβ and p-tau biomarkers is often used for more accurate diagnosis and prediction of AD progression. Along with AD being a neurodegenerative disease, it is associated with other chronic conditions such as cardiovascular disease, obesity, depression, and diabetes because studies have shown that these comorbid conditions make people more vulnerable to AD. In the first part of this review, we discuss that biofluid-based biomarkers such as Aβ, p-Tau in cerebrospinal fluid (CSF) and Aβ & p-Tau in plasma could be used as an alternative sensitive technique to diagnose AD. In the second part, we discuss the underlying molecular mechanisms of chronic conditions linked with AD and how they affect the patients in clinical care.
Collapse
Affiliation(s)
| | - Javaria Baig
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sudhir Kshirsagar
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Neurology Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
5
|
Li W, Li JY. Overlaps and divergences between tauopathies and synucleinopathies: a duet of neurodegeneration. Transl Neurodegener 2024; 13:16. [PMID: 38528629 DOI: 10.1186/s40035-024-00407-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/28/2024] [Indexed: 03/27/2024] Open
Abstract
Proteinopathy, defined as the abnormal accumulation of proteins that eventually leads to cell death, is one of the most significant pathological features of neurodegenerative diseases. Tauopathies, represented by Alzheimer's disease (AD), and synucleinopathies, represented by Parkinson's disease (PD), show similarities in multiple aspects. AD manifests extrapyramidal symptoms while dementia is also a major sign of advanced PD. We and other researchers have sequentially shown the cross-seeding phenomenon of α-synuclein (α-syn) and tau, reinforcing pathologies between synucleinopathies and tauopathies. The highly overlapping clinical and pathological features imply shared pathogenic mechanisms between the two groups of disease. The diagnostic and therapeutic strategies seemingly appropriate for one distinct neurodegenerative disease may also apply to a broader spectrum. Therefore, a clear understanding of the overlaps and divergences between tauopathy and synucleinopathy is critical for unraveling the nature of the complicated associations among neurodegenerative diseases. In this review, we discuss the shared and diverse characteristics of tauopathies and synucleinopathies from aspects of genetic causes, clinical manifestations, pathological progression and potential common therapeutic approaches targeting the pathology, in the aim to provide a timely update for setting the scheme of disease classification and provide novel insights into the therapeutic development for neurodegenerative diseases.
Collapse
Affiliation(s)
- Wen Li
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, China Medical University, Shenyang, 110122, China
| | - Jia-Yi Li
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, China Medical University, Shenyang, 110122, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, BMC A10, 22184, Lund, Sweden.
| |
Collapse
|
6
|
Sen A, Avsar O, Eliacik S, Uysal Tan F. Association between Alzheimer's disease, MAPT gene mutation and some biochemical biomarkers. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:1139-1148. [PMID: 38319996 DOI: 10.1080/15257770.2024.2313573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024]
Abstract
Alzheimer's Disease (AD) is a multifactorial neurodegenerative disease and there is still no definitive treatment today. Early diagnosis of the disease is important, but there are almost no biomarkers that can be used in early diagnosis. The cerebrospinal fluid used in the diagnosis of the disease is not sufficient and is very difficult to obtain. Therefore, blood biomarkers that are less costly, less invasive, easily accessible, and can be used in long-term studies would be a better alternative. The aim of this study is to determine the relationship between Alzheimer's Disease and P301L MAPT gene mutation, homocysteine, folate and uric acid. 101 Alzheimer's patients and 101 healthy individuals were included in this study. Mutation analysis was performed using the Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP) method with blood samples taken from the subjects. There was no significant difference between the patient and control groups in terms of homocysteine (p = 0.771), folate (p = 0.366) and uric acid (p = 0.860). When the genotypes were compared between the patient and control groups in terms of MAPT gene mutation (P301L), no statistically significant difference was detected (p = 0.081). There are very few studies in the literature investigating the relationship between Alzheimer's disease and P301L MAPT gene mutation. Additionally, there is no study investigating the relationship between Alzheimer's disease and homocysteine, folate, uric acid and P301L MAPT mutation in the Turkish population. We believe that this study has shed light on future studies.
Collapse
Affiliation(s)
- Aysenur Sen
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Hitit University, Corum, Türkiye
| | - Orcun Avsar
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Hitit University, Corum, Türkiye
| | - Sinan Eliacik
- Department of Neurology, Faculty of Medicine, Hitit University, Corum, Türkiye
| | - Funda Uysal Tan
- Department of Neurology, Faculty of Medicine, Hitit University, Corum, Türkiye
| |
Collapse
|
7
|
Bali S, Singh R, Wydorski PM, Wosztyl A, Perez VA, Chen D, Rizo J, Joachimiak LA. Ensemble-based design of tau to inhibit aggregation while preserving biological activity. RESEARCH SQUARE 2024:rs.3.rs-3796916. [PMID: 38313287 PMCID: PMC10836093 DOI: 10.21203/rs.3.rs-3796916/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The microtubule-associated protein tau is implicated in neurodegenerative diseases characterized by amyloid formation. Mutations associated with frontotemporal dementia increase tau aggregation propensity and disrupt its endogenous microtubule-binding activity. The structural relationship between aggregation propensity and biological activity remains unclear. We employed a multi-disciplinary approach, including computational modeling, NMR, cross-linking mass spectrometry, and cell models to design tau sequences that stabilize its structural ensemble. Our findings reveal that substitutions near the conserved 'PGGG' beta-turn motif can modulate local conformation, more stably engaging in interactions with the 306VQIVYK311 amyloid motif to decrease aggregation in vitro and in cells. Designed tau sequences maintain microtubule binding and explain why 3R isoforms of tau exhibit reduced pathogenesis over 4R isoforms. We propose a simple mechanism to reduce the formation of pathogenic species while preserving biological function, offering insights for therapeutic strategies aimed at reducing protein misfolding in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sofia Bali
- Molecular Biophysics Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas
| | - Ruhar Singh
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas
| | - Pawel M Wydorski
- Molecular Biophysics Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas
| | - Aleksandra Wosztyl
- Molecular Biophysics Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Valerie A Perez
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas
| | - Dailu Chen
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States Southwestern Medical Center, Dallas, TX 75390, United States
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Lukasz A Joachimiak
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| |
Collapse
|
8
|
Taylor LW, Simzer EM, Pimblett C, Lacey-Solymar OTT, McGeachan RI, Meftah S, Rose JL, Spires-Jones MP, Holt K, Catterson JH, Koch H, Liaquat I, Clarke JH, Skidmore J, Smith C, Booker SA, Brennan PM, Spires-Jones TL, Durrant CS. p-tau Ser356 is associated with Alzheimer's disease pathology and is lowered in brain slice cultures using the NUAK inhibitor WZ4003. Acta Neuropathol 2024; 147:7. [PMID: 38175261 PMCID: PMC10766794 DOI: 10.1007/s00401-023-02667-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Tau hyperphosphorylation and aggregation is a common feature of many dementia-causing neurodegenerative diseases. Tau can be phosphorylated at up to 85 different sites, and there is increasing interest in whether tau phosphorylation at specific epitopes, by specific kinases, plays an important role in disease progression. The AMP-activated protein kinase (AMPK)-related enzyme NUAK1 has been identified as a potential mediator of tau pathology, whereby NUAK1-mediated phosphorylation of tau at Ser356 prevents the degradation of tau by the proteasome, further exacerbating tau hyperphosphorylation and accumulation. This study provides a detailed characterisation of the association of p-tau Ser356 with progression of Alzheimer's disease pathology, identifying a Braak stage-dependent increase in p-tau Ser356 protein levels and an almost ubiquitous presence in neurofibrillary tangles. We also demonstrate, using sub-diffraction-limit resolution array tomography imaging, that p-tau Ser356 co-localises with synapses in AD postmortem brain tissue, increasing evidence that this form of tau may play important roles in AD progression. To assess the potential impacts of pharmacological NUAK inhibition in an ex vivo system that retains multiple cell types and brain-relevant neuronal architecture, we treated postnatal mouse organotypic brain slice cultures from wildtype or APP/PS1 littermates with the commercially available NUAK1/2 inhibitor WZ4003. Whilst there were no genotype-specific effects, we found that WZ4003 results in a culture-phase-dependent loss of total tau and p-tau Ser356, which corresponds with a reduction in neuronal and synaptic proteins. By contrast, application of WZ4003 to live human brain slice cultures results in a specific lowering of p-tau Ser356, alongside increased neuronal tubulin protein. This work identifies differential responses of postnatal mouse organotypic brain slice cultures and adult human brain slice cultures to NUAK1 inhibition that will be important to consider in future work developing tau-targeting therapeutics for human disease.
Collapse
Affiliation(s)
- Lewis W Taylor
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Elizabeth M Simzer
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Claire Pimblett
- Centre for Discovery Brain Sciences, 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
| | - Soraya Meftah
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jamie L Rose
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | | | - Kristján Holt
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - James H Catterson
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Henner Koch
- Department of Neurology, Epileptology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Imran Liaquat
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Jonathan H Clarke
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| | - John Skidmore
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| | - Colin Smith
- The Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Sam A Booker
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, The University of Edinburgh, Edinburgh, UK
| | - Paul M Brennan
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
- The Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Brain Tumour Centre of Excellence, CRUK Edinburgh Centre, 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
| | - Claire S Durrant
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK.
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
9
|
Sun K, Patel T, Kang SG, Yarahmady A, Srinivasan M, Julien O, Heras J, Mok SA. Disease-Associated Mutations in Tau Encode for Changes in Aggregate Structure Conformation. ACS Chem Neurosci 2023; 14:4282-4297. [PMID: 38054595 PMCID: PMC10741665 DOI: 10.1021/acschemneuro.3c00422] [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: 06/17/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
The accumulation of tau fibrils is associated with neurodegenerative diseases, which are collectively termed tauopathies. Cryo-EM studies have shown that the packed fibril core of tau adopts distinct structures in different tauopathies, such as Alzheimer's disease, corticobasal degeneration, and progressive supranuclear palsy. A subset of tauopathies are linked to missense mutations in the tau protein, but it is not clear whether these mutations impact the structure of tau fibrils. To answer this question, we developed a high-throughput protein purification platform and purified a panel of 37 tau variants using the full-length 0N4R splice isoform. Each of these variants was used to create fibrils in vitro, and their relative structures were studied using a high-throughput protease sensitivity platform. We find that a subset of the disease-associated mutations form fibrils that resemble wild-type tau, while others are strikingly different. The impact of mutations on tau structure was not clearly associated with either the location of the mutation or the relative kinetics of fibril assembly, suggesting that tau mutations alter the packed core structures through a complex molecular mechanism. Together, these studies show that single-point mutations can impact the assembly of tau into fibrils, providing insight into its association with pathology and disease.
Collapse
Affiliation(s)
- Kerry
T. Sun
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Tark Patel
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Sang-Gyun Kang
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Allan Yarahmady
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Mahalashmi Srinivasan
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Jónathan Heras
- Department
of Mathematics and Computer Sciences, University
of La Rioja, Logroño, Spain 26004
| | - Sue-Ann Mok
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| |
Collapse
|
10
|
Paterno G, Bell BM, Riley-DiPaolo A, LaVoie MJ, Giasson BI. Polymerization of recombinant tau core fragments in vitro and seeding studies in cultured cells. Front Neurosci 2023; 17:1268360. [PMID: 38161790 PMCID: PMC10757379 DOI: 10.3389/fnins.2023.1268360] [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: 07/27/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
The relative polymerization of specific tau protein cores that define Alzheimer's disease, Pick's disease and corticobasal degeneration were investigated using amyloid fluorometry and electron microscopy. In addition, the relative prion-like activities of polymers comprised of these respective tau protein segments were investigated in a cell-based assay. It is demonstrated that the seeding activities of specific tau core fibrils are affected by the presence of pathogenic tau missense mutations and the microtubule binding domain composition of tau. The unique impact of tau phosphorylation on seeding propensity was also investigated by altering stretches of phospho-mimetic and phospho-null residues in the presence of Alzheimer's disease tau core fibrils. These results have important mechanistic implications for mutation and isoform-specific driven pathogenesis.
Collapse
Affiliation(s)
- Giavanna Paterno
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Brach M. Bell
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Alexis Riley-DiPaolo
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Matthew J. LaVoie
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Benoit I. Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
11
|
Bali S, Singh R, Wydorski PM, Wosztyl A, Perez VA, Chen D, Rizo J, Joachimiak LA. Ensemble-based design of tau to inhibit aggregation while preserving biological activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571598. [PMID: 38168322 PMCID: PMC10760154 DOI: 10.1101/2023.12.13.571598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The microtubule-associated protein tau is implicated in neurodegenerative diseases characterized by amyloid formation. Mutations associated with frontotemporal dementia increase tau aggregation propensity and disrupt its endogenous microtubule-binding activity. The structural relationship between aggregation propensity and biological activity remains unclear. We employed a multi-disciplinary approach, including computational modeling, NMR, cross-linking mass spectrometry, and cell models to design tau sequences that stabilize its structural ensemble. Our findings reveal that substitutions near the conserved 'PGGG' beta-turn motif can modulate local conformation, more stably engaging in interactions with the 306 VQIVYK 311 amyloid motif to decrease aggregation in vitro and in cells. Designed tau sequences maintain microtubule binding and explain why 3R isoforms of tau exhibit reduced pathogenesis over 4R isoforms. We propose a simple mechanism to reduce the formation of pathogenic species while preserving biological function, offering insights for therapeutic strategies aimed at reducing protein misfolding in neurodegenerative diseases.
Collapse
|
12
|
Smith ED, Vo Q, Giasson BI, Borchelt DR, Prokop S, Chakrabarty P. Human tauopathy strains defined by phosphorylation in R1-R2 repeat domains of tau. Acta Neuropathol Commun 2023; 11:172. [PMID: 37891635 PMCID: PMC10612232 DOI: 10.1186/s40478-023-01664-0] [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: 08/05/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Distinctive post-translational modifications (PTM) characterize tau inclusions found in tauopathy patients. Using detergent-insoluble tau isolated from Alzheimer's disease (AD-tau) or Progressive Supranuclear Palsy (PSP-tau) patients, we provide insights into whether phosphorylation of critical residues determine templated tau seeding. Our initial data with phosphorylation-ablating mutations (Ser/Thr → Ala) on select sites of P301L tau showed no changes in seeding efficacy by AD-tau or PSP-tau. Interestingly, when specific sites in the R1-R2 repeat domains (Ser262/Thr263/Ser289/Ser305) were mutated to phosphorylation-mimicking amino acid Glu, it substantially reduced the seeding efficiency of AD-tau, but not PSP-tau seeds. The resultant detergent-insoluble tau shows deficient phosphorylation on AT8, AT100, AT180 and PHF1 epitopes, indicating inter-domain cooperativity. We further identify Ser305 as a critical determinant of AD-tau-specific seeding, whereby the phospho-mimicking Ser305Glu tau abrogates seeding by AD-tau but not PSP-tau. This suggests that phosphorylation on Ser305 could be related to the formation of disease-specific tau strains. Our results highlight the existence of a phospho-PTM code in tau seeding and further demonstrate the distinctive nature of this code in 4R tauopathies.
Collapse
Affiliation(s)
- Ethan D Smith
- Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BMS J484, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Quan Vo
- Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BMS J484, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Benoit I Giasson
- Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BMS J484, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - David R Borchelt
- Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BMS J484, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BMS J484, Gainesville, FL, 32610, USA
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32610, USA
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Disease, University of Florida, 1275 Center Drive, BMS J484, Gainesville, FL, 32610, USA.
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
13
|
Paterno G, Torrellas J, Bell BM, Gorion KMM, Quintin SS, Hery GP, Prokop S, Giasson BI. Novel Conformation-Dependent Tau Antibodies Are Modulated by Adjacent Phosphorylation Sites. Int J Mol Sci 2023; 24:13676. [PMID: 37761979 PMCID: PMC10530490 DOI: 10.3390/ijms241813676] [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: 07/18/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Tau proteins within the adult central nervous system (CNS) are found to be abnormally aggregated into heterogeneous filaments in neurodegenerative diseases, termed tauopathies. These tau inclusions are pathological hallmarks of Alzheimer's disease (AD), Pick's disease (PiD), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP). The neuropathological hallmarks of these diseases burden several cell types within the CNS, and have also been shown to be abundantly phosphorylated. The mechanism(s) by which tau aggregates in the CNS is not fully known, but it is hypothesized that hyperphosphorylated tau may precede and further promote filament formation, leading to the production of these pathological inclusions. In the studies herein, we generated and thoroughly characterized two novel conformation-dependent tau monoclonal antibodies that bind to residues Pro218-Glu222, but are sensitive to denaturing conditions and highly modulated by adjacent downstream phosphorylation sites. These epitopes are present in the neuropathological hallmarks of several tauopathies, including AD, PiD, CBD, and PSP. These novel antibodies will further enable investigation of tau-dependent pathological inclusion formation and enhance our understanding of the phosphorylation signatures within tauopathies with the possibility of new biomarker developments.
Collapse
Affiliation(s)
- Giavanna Paterno
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Jose Torrellas
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Brach M. Bell
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Kimberly-Marie M. Gorion
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Stephan S. Quintin
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Gabriela P. Hery
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benoit I. Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
14
|
Xia Y, Bell BM, Kim JD, Giasson BI. Tau mutation S356T in the three repeat isoform leads to microtubule dysfunction and promotes prion-like seeded aggregation. Front Neurosci 2023; 17:1181804. [PMID: 37304025 PMCID: PMC10248064 DOI: 10.3389/fnins.2023.1181804] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/03/2023] [Indexed: 06/13/2023] Open
Abstract
Tauopathies are a group of neurodegenerative diseases, which include frontotemporal dementia (FTD) and Alzheimer's disease (AD), broadly defined by the development of tau brain aggregates. Both missense and splicing tau mutations can directly cause early onset FTD. Tau protein is a microtubule-associated protein that stabilizes and regulates microtubules, but this function can be disrupted in disease states. One contributing factor is the balance of different tau isoforms, which can be categorized into either three repeat (3R) or four repeat (4R) isoforms based on the number of microtubule-binding repeats that are expressed. Imbalance of 3R and 4R isoforms in either direction can cause FTD and neurodegeneration. There is also increasing evidence that 3R tauopathies such as Pick's disease form tau aggregates predominantly comprised of 3R isoforms and these can present differently from 4R and mixed 3R/4R tauopathies. In this study, multiple mutations in 3R tau were assessed for MT binding properties and prion-like aggregation propensity. Different missense tau mutations showed varying effects on MT binding depending on molecular location and properties. Of the mutations that were surveyed, S356T tau is uniquely capable of prion-like seeded aggregation and forms extensive Thioflavin positive aggregates. This unique prion-like tau strain will be useful to model 3R tau aggregation and will contribute to the understanding of diverse presentations of different tauopathies.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Brach M. Bell
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Justin D. Kim
- Department of Internal Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Benoit I. Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Xia Y, Bell BM, Giasson BI. Tau Lysine Pseudomethylation Regulates Microtubule Binding and Enhances Prion-like Tau Aggregation. Int J Mol Sci 2023; 24:ijms24098286. [PMID: 37175990 PMCID: PMC10179186 DOI: 10.3390/ijms24098286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/25/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Alzheimer's disease (AD) and frontotemporal dementia (FTD) can be classified as tauopathies, which are a group of neurodegenerative diseases that develop toxic tau aggregates in specific brain regions. These pathological tau inclusions are altered by various post-translational modifications (PTMs) that include phosphorylation, acetylation, and methylation. Tau methylation has emerged as a target of interest for its potential involvement in tau pathomechanisms. Filamentous tau aggregates isolated from patients with AD are methylated at multiple lysine residues, although the exact methyltransferases have not been identified. One strategy to study the site-specific effects of methylation is to create methylation mimetics using a KFC model, which replaces lysine (K) with a hydrophobic group such as phenylalanine (F) to approximate the effects of lysine methylation (C or methyl group). In this study, tau methylmimetics were used to model several functional aspects of tau methylation such as effects on microtubule binding and tau aggregation in cell models. Overall, several tau methylmimetics displayed impaired microtubule binding, and tau methylmimetics enhanced prion-like seeded aggregation in the context of the FTD tau mutation P301L. Like other PTMs, tau methylation is a contributing factor to tau pathogenesis and could be a potential therapeutic drug target for the treatment of different tauopathies.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brach M Bell
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
16
|
Xia Y, Prokop S, Bell BM, Gorion KMM, Croft CL, Nasif L, Xu G, Riffe CJ, Manaois AN, Strang KH, Quintin SS, Paterno G, Tansey MG, Borchelt DR, Golde TE, Giasson BI. Pathogenic tau recruits wild-type tau into brain inclusions and induces gut degeneration in transgenic SPAM mice. Commun Biol 2022; 5:446. [PMID: 35550593 PMCID: PMC9098443 DOI: 10.1038/s42003-022-03373-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/14/2022] [Indexed: 01/04/2023] Open
Abstract
Pathological tau inclusions are neuropathologic hallmarks of many neurodegenerative diseases. We generated and characterized a transgenic mouse model expressing pathogenic human tau with S320F and P301S aggregating mutations (SPAM) at transgene levels below endogenous mouse tau protein levels. This mouse model develops a predictable temporal progression of tau pathology in the brain with biochemical and ultrastructural properties akin to authentic tau inclusions. Surprisingly, pathogenic human tau extensively recruited endogenous mouse tau into insoluble aggregates. Despite the early onset and rapid progressive nature of tau pathology, major neuroinflammatory and transcriptional changes were only detectable at later time points. Moreover, tau SPAM mice are the first model to develop loss of enteric neurons due to tau accumulation resulting in a lethal phenotype. With moderate transgene expression, rapidly progressing tau pathology, and a highly predictable lethal phenotype, the tau SPAM model reveals new associations of tau neurotoxicity in the brain and intestinal tract.
Collapse
Affiliation(s)
- Yuxing Xia
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Stefan Prokop
- grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Department of Pathology, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Brach M. Bell
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Kimberly-Marie M. Gorion
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Cara L. Croft
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Lith Nasif
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Guilian Xu
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Cara J. Riffe
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Alyssa N. Manaois
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Kevin H. Strang
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Stephan S. Quintin
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Giavanna Paterno
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Malú Gámez Tansey
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - David R. Borchelt
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Todd E. Golde
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Benoit I. Giasson
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| |
Collapse
|
17
|
Zhang Y, Wu KM, Yang L, Dong Q, Yu JT. Tauopathies: new perspectives and challenges. Mol Neurodegener 2022; 17:28. [PMID: 35392986 PMCID: PMC8991707 DOI: 10.1186/s13024-022-00533-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tauopathies are a class of neurodegenerative disorders characterized by neuronal and/or glial tau-positive inclusions. MAIN BODY Clinically, tauopathies can present with a range of phenotypes that include cognitive/behavioral-disorders, movement disorders, language disorders and non-specific amnestic symptoms in advanced age. Pathologically, tauopathies can be classified based on the predominant tau isoforms that are present in the inclusion bodies (i.e., 3R, 4R or equal 3R:4R ratio). Imaging, cerebrospinal fluid (CSF) and blood-based tau biomarkers have the potential to be used as a routine diagnostic strategy and in the evaluation of patients with tauopathies. As tauopathies are strongly linked neuropathologically and genetically to tau protein abnormalities, there is a growing interest in pursuing of tau-directed therapeutics for the disorders. Here we synthesize emerging lessons on tauopathies from clinical, pathological, genetic, and experimental studies toward a unified concept of these disorders that may accelerate the therapeutics. CONCLUSIONS Since tauopathies are still untreatable diseases, efforts have been made to depict clinical and pathological characteristics, identify biomarkers, elucidate underlying pathogenesis to achieve early diagnosis and develop disease-modifying therapies.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Liu Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| |
Collapse
|
18
|
De Luigi A, Colombo L, Russo L, Ricci C, Bastone A, Cimini S, Tagliavini F, Rossi G, Cantù L, Del Favero E, Salmona M. Biochemical and biophysical features of disease-associated tau mutants V363A and V363I. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140755. [PMID: 34999006 DOI: 10.1016/j.bbapap.2022.140755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/07/2021] [Accepted: 01/01/2022] [Indexed: 06/14/2023]
Abstract
The comprehension of pathogenetic mechanisms in tauopathy-associated neurodegenerative diseases can be improved by the knowledge of the biochemical and biophysical features of mutated tau proteins. Here, we used the full-length, wild-type tau, the V363A and V363I mutated species, associated with pathology, and the P301L mutated tau as a benchmark. Using several techniques, including small-angle X-ray scattering, atomic force microscopy, thioflavin T binding, and electrophoretic separation, we compared their course from intrinsically disordered monomers in solution to early-stage recruitment in complexes and then aggregates of increasing size over long periods up to the asymptotic aggregative behavior of full-length tau proteins. We showed that diversity in the kinetics of recruitment and aggregate structure occurs from the beginning and spreads all over their pathway to very large objects. The different extents of conformational changes and types of molecular assemblies among the proteins were also reflected in their in vitro toxicity; this variation could correlate with physiopathology in humans, considering that the P301L mutation is more aggressive than V363A, especially V363I. This study identified the presence of aggregation intermediates and corroborated the oligomeric hypothesis of tauopathies.
Collapse
Affiliation(s)
- Ada De Luigi
- Department of Molecular Biochemistry and Pharmacology, Mario Negri Institute of Pharmacological Research IRCCS, Milano, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, Mario Negri Institute of Pharmacological Research IRCCS, Milano, Italy
| | - Luca Russo
- Department of Molecular Biochemistry and Pharmacology, Mario Negri Institute of Pharmacological Research IRCCS, Milano, Italy
| | - Caterina Ricci
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Antonio Bastone
- Department of Molecular Biochemistry and Pharmacology, Mario Negri Institute of Pharmacological Research IRCCS, Milano, Italy
| | - Sara Cimini
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fabrizio Tagliavini
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giacomina Rossi
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Laura Cantù
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Elena Del Favero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy.
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, Mario Negri Institute of Pharmacological Research IRCCS, Milano, Italy.
| |
Collapse
|
19
|
Tau K321/K353 pseudoacetylation within KXGS motifs regulates tau-microtubule interactions and inhibits aggregation. Sci Rep 2021; 11:17069. [PMID: 34426645 PMCID: PMC8382713 DOI: 10.1038/s41598-021-96627-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease is the leading cause of dementia and a defining hallmark is the progressive brain deposition of tau aggregates. The insidious accumulation of brain tau inclusions is also involved in a group of neurodegenerative diseases termed frontotemporal dementias. In all of these disorders, tau aggregates are enriched in post-translational modifications including acetylation, which has recently been identified at multiple sites. While most evidence suggest that tau acetylation is detrimental and promotes tau aggregation, a few studies support that tau acetylation within the KXGS motif can be protective and inhibit tau aggregation. To model site-specific acetylation at K259, K290, K321, and K353, acetylmimetics were created by mutating lysine to glutamine residues, which approximates size and charge of acetylation. HEK293T cells were transfected to express wild type tau, tau pathogenic mutations (P301L and P301L/S320F) or tau acetylmimetics and assessed by cell-based assays for microtubule binding and tau aggregation. Acetylmimetics within the KXGS motif (K259Q, K290Q, K321Q, K353Q) leads to significant decreased tau-microtubule interactions. Acetylmimetics K321Q and K353Q within the context of the pathogenic P301L tau mutation strongly inhibited prion-like seeded aggregation. This protective effect was confirmed to decrease intrinsic aggregation of P301L/S320F tau double mutation. Surprisingly, K321Q and K353Q acetylmimetics altered the conformational structure of P301L/S320F tau to extensively impair Thioflavin S binding. Site-specific acetylation of tau at K321 and K353 could represent a natural protective mechanism against tau aggregation and could be a potential therapeutic target.
Collapse
|
20
|
Xia Y, Prokop S, Giasson BI. "Don't Phos Over Tau": recent developments in clinical biomarkers and therapies targeting tau phosphorylation in Alzheimer's disease and other tauopathies. Mol Neurodegener 2021; 16:37. [PMID: 34090488 PMCID: PMC8180161 DOI: 10.1186/s13024-021-00460-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Phosphorylation is one of the most prevalent post-translational modifications found in aggregated tau isolated from Alzheimer’s disease (AD) patient brains. In tauopathies like AD, increased phosphorylation or hyperphosphorylation can contribute to microtubule dysfunction and is associated with tau aggregation. In this review, we provide an overview of the structure and functions of tau protein as well as the physiologic roles of tau phosphorylation. We also extensively survey tau phosphorylation sites identified in brain tissue and cerebrospinal fluid from AD patients compared to age-matched healthy controls, which may serve as disease-specific biomarkers. Recently, new assays have been developed to measure minute amounts of specific forms of phosphorylated tau in both cerebrospinal fluid and plasma, which could potentially be useful for aiding clinical diagnosis and monitoring disease progression. Additionally, multiple therapies targeting phosphorylated tau are in various stages of clinical trials including kinase inhibitors, phosphatase activators, and tau immunotherapy. With promising early results, therapies that target phosphorylated tau could be useful at slowing tau hyperphosphorylation and aggregation in AD and other tauopathies.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA.,Department of Pathology, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA. .,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA. .,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA.
| |
Collapse
|
21
|
Ceyzériat K, Zilli T, Millet P, Frisoni GB, Garibotto V, Tournier BB. Learning from the Past: A Review of Clinical Trials Targeting Amyloid, Tau and Neuroinflammation in Alzheimer's Disease. Curr Alzheimer Res 2021; 17:112-125. [PMID: 32129164 DOI: 10.2174/1567205017666200304085513] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 02/11/2020] [Accepted: 03/01/2020] [Indexed: 12/31/2022]
Abstract
Alzheimer's Disease (AD) is the most common neurodegenerative disease and cause of dementia. Characterized by amyloid plaques and neurofibrillary tangles of hyperphosphorylated Tau, AD pathology has been intensively studied during the last century. After a long series of failed trials of drugs targeting amyloid or Tau deposits, currently, hope lies in the positive results of one Phase III trial, highly debated, and on other ongoing trials. In parallel, some approaches target neuroinflammation, another central feature of AD. Therapeutic strategies are initially evaluated on animal models, in which the various drugs have shown effects on the target (decreasing amyloid, Tau and neuroinflammation) and sometimes on cognitive impairment. However, it is important to keep in mind that rodent models have a less complex brain than humans and that the pathology is generally not fully represented. Although they are indispensable tools in the drug discovery process, results obtained from animal models must be viewed with caution. In this review, we focus on the current status of disease-modifying therapies targeting amyloid, Tau and neuroinflammation with particular attention on the discrepancy between positive preclinical results on animal models and failures in clinical trials.
Collapse
Affiliation(s)
- Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland.,Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University and Geneva University Hospitals, Geneva, Switzerland.,Division of Radiation Oncology, Department of Oncology, Geneva University and Geneva University Hospitals, Geneva, Switzerland
| | - Thomas Zilli
- Division of Radiation Oncology, Department of Oncology, Geneva University and Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Giovanni B Frisoni
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University and Geneva University Hospitals, Geneva, Switzerland.,IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University and Geneva University Hospitals, Geneva, Switzerland
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
22
|
Xia Y, Nasif L, Giasson BI. Pathogenic MAPT mutations Q336H and Q336R have isoform-dependent differences in aggregation propensity and microtubule dysfunction. J Neurochem 2021; 158:455-466. [PMID: 33772783 DOI: 10.1111/jnc.15358] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/24/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022]
Abstract
Tauopathies are a group of heterogeneous neurodegenerative disorders characterized by brain deposition of tau inclusions. These insidious disorders include Alzheimer's disease and frontotemporal dementia, the two leading causes of dementia. Mutations in the microtubule-associated protein tau (MAPT) gene lead to familial forms of frontotemporal dementia. Previously, we used cell-based assays to screen over 20 missense tau mutations and found that decreased microtubule (MT) binding affinity was the most shared property. As a break from this trend, the MAPT mutations Q336H and Q336R are thought to promote MT assembly rather than inhibit it based on in vitro studies. Q336H and Q336R MAPT mutations also cause early onset frontotemporal dementia with Pick bodies, which are mostly composed of 3R tau isoforms. To provide further insights on the pathobiology of these mutations, we assessed Q336H and Q336R tau mutants for aggregation propensity and MT binding in cell-based assays in the context of both 0N3R and 0N4R tau isoforms. Q336R tau was prone to prion-like seeded aggregation but both Q336H and Q336R tau led to increased MT binding. Additionally, we found that different tau isoforms with these mutations heterogeneously regulate different MT subpopulations of tyrosinated and acetylated MTs, markers of newly formed MTs and stable MTs. The Q336H and Q336R tau mutations may exemplify an alternative mechanism where pathogenic tau can bind MTs with higher affinity and hyperstabilize MTs, which prevent proper MT regulation and homeostasis.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lith Nasif
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
23
|
Xia Y, Chen Z, Xu G, Borchelt DR, Ayers JI, Giasson BI. Novel SOD1 monoclonal antibodies against the electrostatic loop preferentially detect misfolded SOD1 aggregates. Neurosci Lett 2020; 742:135553. [PMID: 33346076 DOI: 10.1016/j.neulet.2020.135553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurological disease that leads to motor neuron degeneration and paralysis. Superoxide dismutase (SOD1) mutations are the second most common cause of familial ALS and are responsible for up to 20 % of familial ALS cases. In ALS patients, SOD1 can form toxic misfolded aggregates that deposit in the brain and spinal cord. To better detect SOD1 aggregates and expand the repertoire of conformational SOD1 antibodies, SOD1 monoclonal antibodies were generated by immunizing SOD1 knockout mice with an SOD1 fragment consisting of amino acids 129-146, which make up part of the electrostatic loop. A series of hybridomas secreting antibodies were screened and five different SOD1 monoclonal antibodies (2C10, 2F8, 4B11, 5H5, and 5A10) were found to preferentially detect denatured or aggregated SOD1 by enzyme-linked immunosorbent assay (ELISA), filter trap assay, and immunohistochemical analysis in SOD1 mouse models. The staining with these antibodies was compared to Campbell-Switzer argyrophilic reactivity of pathological inclusions. These new conformational selective SOD1 antibodies will be useful for clinical diagnosis of SOD1 ALS and potentially for passive immunotherapy.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zhijuan Chen
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Guilian Xu
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - David R Borchelt
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jacob I Ayers
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA; Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
24
|
Muralidar S, Ambi SV, Sekaran S, Thirumalai D, Palaniappan B. Role of tau protein in Alzheimer's disease: The prime pathological player. Int J Biol Macromol 2020; 163:1599-1617. [PMID: 32784025 DOI: 10.1016/j.ijbiomac.2020.07.327] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is a prevalently found tauopathy characterized by memory loss and cognitive insufficiency. AD is an age-related neurodegenerative disease with two major hallmarks which includes extracellular amyloid plaques made of amyloid-β (Aβ) and intracellular neurofibrillary tangles of hyperphosphorylated tau. With population aging worldwide, there is an indispensable need for treatment strategies that can potentially manage this developing dementia. Despite broad researches on targeting Aβ in the past two decades, research findings on Aβ targeted therapeutics failed to prove efficacy in the treatment of AD. Tau protein with its extensive pathological role in several neurodegenerative diseases can be considered as a promising target candidate for developing therapeutic interventions. The abnormal hyperphosphorylation of tau plays detrimental pathological functions which ultimately lead to neurodegeneration. This review will divulge the importance of tau in AD pathogenesis, the interplay of Aβ and tau, the pathological functions of tau, and potential therapeutic strategies for an effective management of neuronal disorders.
Collapse
Affiliation(s)
- Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India.
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India; Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Diraviyam Thirumalai
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Balamurugan Palaniappan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| |
Collapse
|
25
|
Xia Y, Prokop S, Gorion KMM, Kim JD, Sorrentino ZA, Bell BM, Manaois AN, Chakrabarty P, Davies P, Giasson BI. Tau Ser208 phosphorylation promotes aggregation and reveals neuropathologic diversity in Alzheimer's disease and other tauopathies. Acta Neuropathol Commun 2020; 8:88. [PMID: 32571418 PMCID: PMC7310041 DOI: 10.1186/s40478-020-00967-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Tau protein abnormally aggregates in tauopathies, a diverse group of neurologic diseases that includes Alzheimer’s disease (AD). In early stages of disease, tau becomes hyperphosphorylated and mislocalized, which can contribute to its aggregation and toxicity. We demonstrate that tau phosphorylation at Ser208 (pSer208) promotes microtubule dysfunction and tau aggregation in cultured cells. Comparative assessment of the epitopes recognized by antibodies AT8, CP13, and 7F2 demonstrates that CP13 and 7F2 are specific for tau phosphorylation at Ser202 and Thr205, respectively, independently of the phosphorylation state of adjacent phosphorylation sites. Supporting the involvement of pSer208 in tau pathology, a novel monoclonal antibody 3G12 specific for tau phosphorylation at Ser208 revealed strong reactivity of tau inclusions in the brains of PS19 and rTg4510 transgenic mouse models of tauopathy. 3G12 also labelled neurofibrillary tangles in brains of patients with AD but revealed differential staining compared to CP13 and 7F2 for other types of tau pathologies such as in neuropil threads and neuritic plaques in AD, tufted astrocytes in progressive supranuclear palsy and astrocytic plaques in corticobasal degeneration. These results support the hypothesis that tau phosphorylation at Ser208 strongly contributes to unique types of tau aggregation and may be a reliable marker for the presence of mature neurofibrillary tangles.
Collapse
|