151
|
Lantero‐Rodriguez J, Snellman A, Benedet AL, Milà‐Alomà M, Camporesi E, Montoliu‐Gaya L, Ashton NJ, Vrillon A, Karikari TK, Gispert JD, Salvadó G, Shekari M, Toomey CE, Lashley TL, Zetterberg H, Suárez‐Calvet M, Brinkmalm G, Rosa Neto P, Blennow K. P-tau235: a novel biomarker for staging preclinical Alzheimer's disease. EMBO Mol Med 2021; 13:e15098. [PMID: 34725927 PMCID: PMC8649868 DOI: 10.15252/emmm.202115098] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 12/05/2022] Open
Abstract
Alzheimer's disease (AD) is characterised by a long preclinical phase. Although phosphorylated tau (p-tau) species such as p-tau217 and p-tau231 provide accurate detection of early pathological changes, other biomarkers capable of staging disease progression during preclinical AD are still needed. Combining exploratory and targeted mass spectrometry methods in neuropathologically confirmed brain tissue, we observed that p-tau235 is a prominent feature of AD pathology. In addition, p-tau235 seemed to be preceded by p-tau231, in what appeared to be a sequential phosphorylation event. To exploit its biomarker potential in cerebrospinal fluid (CSF), we developed and validated a new p-tau235 Simoa assay. Using three clinical cohorts, we demonstrated that (i) CSF p-235 increases early in AD continuum, and (ii) changes in CSF p-tau235 and p-tau231 levels during preclinical AD are consistent with the sequential phosphorylation evidence in AD brain. In conclusion, CSF p-tau235 appears to be not only a highly specific biomarker of AD but also a promising staging biomarker for the preclinical phase. Thus, it could prove useful tracking disease progression and help enriching clinical trial recruitment.
Collapse
|
152
|
Leslie SN, Kanyo J, Datta D, Wilson RS, Zeiss C, Duque A, Lam TT, Arnsten AFT, Nairn AC. Simple, Single-Shot Phosphoproteomic Analysis of Heat-Stable Tau Identifies Age-Related Changes in pS235- and pS396-Tau Levels in Non-human Primates. Front Aging Neurosci 2021; 13:767322. [PMID: 34867294 PMCID: PMC8637411 DOI: 10.3389/fnagi.2021.767322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Age is the most significant risk factor for Alzheimer's disease (AD), and understanding its role in specific aspects of AD pathology will be critical for therapeutic development. Neurofibrillary tangles composed of hyperphosphorylated tau are a quintessential hallmark of AD. To study age-related changes in tau phosphorylation, we developed a simple, antibody-free approach for single shot analysis of tau phosphorylation across the entire protein by liquid-chromatography tandem mass spectrometry. This methodology is species independent; thus, while initially developed in a rodent model, we utilized this technique to analyze 36 phosphorylation sites on rhesus monkey tau from the prefrontal cortex (PFC), a region vulnerable to AD-linked degeneration. Data are available via ProteomeXchange with identifier PXD027971. We identified novel, age-related changes in tau phosphorylation in the rhesus monkey PFC and analyzed patterns of phosphorylation change across domains of the protein. We confirmed a significant increase and positive correlation with age of phosphorylated serine 235 tau and phosphorylated serine 396 tau levels in an expanded cohort of 14 monkeys. Histology showed robust labeling for tau phosphorylated at these sites in vulnerable layer III pyramidal cells in the PFC. The results presented in this study suggest an important role of the natural aging process in tau phosphorylation in rhesus monkey.
Collapse
Affiliation(s)
- Shannon N. Leslie
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT, United States
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Jean Kanyo
- Keck MS & Proteomics Resource, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Dibyadeep Datta
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Rashaun S. Wilson
- Keck MS & Proteomics Resource, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Caroline Zeiss
- Department of Comparative Medicine, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Alvaro Duque
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - TuKiet T. Lam
- Keck MS & Proteomics Resource, Yale School of Medicine, Yale University, New Haven, CT, United States
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, United States
| | - Amy F. T. Arnsten
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Angus C. Nairn
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
153
|
Limorenko G, Lashuel HA. To target Tau pathologies, we must embrace and reconstruct their complexities. Neurobiol Dis 2021; 161:105536. [PMID: 34718129 DOI: 10.1016/j.nbd.2021.105536] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 10/20/2022] Open
Abstract
The accumulation of hyperphosphorylated fibrillar Tau aggregates in the brain is one of the defining hallmarks of Tauopathy diseases, including Alzheimer's disease. However, the primary events or molecules responsible for initiation of the pathological Tau aggregation and spreading remain unknown. The discovery of heparin as an effective inducer of Tau aggregation in vitro was instrumental to enabling different lines of research into the role of Tau aggregation in the pathogenesis of Tauopathies. However, recent proteomics and cryogenic electron microscopy (cryo-EM) studies have revealed that heparin-induced Tau fibrils generated in vitro do not reproduce the biochemical and ultrastructural properties of disease-associated brain-derived Tau fibrils. These observations demand that we reassess our current approaches for investigating the mechanisms underpinning Tau aggregation and pathology formation. Our review article presents an up-to-date survey and analyses of 1) the evolution of our understanding of the interactions between Tau and heparin, 2) the various structural and mechanistic models of the heparin-induced Tau aggregation, 3) the similarities and differences between brain-derived and heparin-induced Tau fibrils; and 4) emerging concepts on the biochemical and structural determinants underpinning Tau pathological heterogeneity in Tauopathies. Our analyses identify specific knowledge gaps and call for 1) embracing the complexities of Tau pathologies; 2) reassessment of current approaches to investigate, model and reproduce pathological Tau aggregation as it occurs in the brain; 3) more research towards a better understanding of the naturally-occurring cofactor molecules that are associated with Tau brain pathology initiation and propagation; and 4) developing improved approaches for in vitro production of the Tau aggregates and fibrils that recapitulate and/or amplify the biochemical and structural complexity and diversity of pathological Tau in Tauopathies. This will result in better and more relevant tools, assays, and mechanistic models, which could significantly improve translational research and the development of drugs and antibodies that have higher chances for success in the clinic.
Collapse
Affiliation(s)
- Galina Limorenko
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
154
|
Eisenbaum M, Pearson A, Gratkowski A, Mouzon B, Mullan M, Crawford F, Ojo J, Bachmeier C. Influence of traumatic brain injury on extracellular tau elimination at the blood-brain barrier. Fluids Barriers CNS 2021; 18:48. [PMID: 34702292 PMCID: PMC8549249 DOI: 10.1186/s12987-021-00283-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 03/14/2023] Open
Abstract
Repetitive head trauma has been associated with the accumulation of tau species in the brain. Our prior work showed brain vascular mural cells contribute to tau processing in the brain, and that these cells progressively degenerate following repetitive mild traumatic brain injury (r-mTBI). The current studies investigated the role of the cerebrovasculature in the elimination of extracellular tau from the brain, and the influence of r-mTBI on these processes. Following intracranial injection of biotin-labeled tau, the levels of exogenous labeled tau residing in the brain were elevated in a mouse model of r-mTBI at 12 months post-injury compared to r-sham mice, indicating reduced tau elimination from the brain following head trauma. This may be the result of decreased caveolin-1 mediated tau efflux at the blood–brain barrier (BBB), as the caveolin inhibitor, methyl-β-cyclodextrin, significantly reduced tau uptake in isolated cerebrovessels and significantly decreased the basolateral-to-apical transit of tau across an in vitro model of the BBB. Moreover, we found that the upstream regulator of endothelial caveolin-1, Mfsd2a, was elevated in r-mTBI cerebrovessels compared to r-sham, which coincided with a decreased expression of cerebrovascular caveolin-1 in the chronic phase following r-mTBI (> 3 months post-injury). Lastly, angiopoietin-1, a mural cell-derived protein governing endothelial Mfsd2a expression, was secreted from r-mTBI cerebrovessels to a greater extent than r-sham animals. Altogether, in the chronic phase post-injury, release of angiopoietin-1 from degenerating mural cells downregulates caveolin-1 expression in brain endothelia, resulting in decreased tau elimination across the BBB, which may describe the accumulation of tau species in the brain following head trauma.
Collapse
Affiliation(s)
- Maxwell Eisenbaum
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA. .,The Open University, Milton Keynes, UK.
| | - Andrew Pearson
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK
| | - Arissa Gratkowski
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Benoit Mouzon
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK.,James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK.,James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Joseph Ojo
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK
| | - Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK.,Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
155
|
Leuzy A, Janelidze S, Mattsson-Carlgren N, Palmqvist S, Jacobs D, Cicognola C, Stomrud E, Vanmechelen E, Dage JL, Hansson O. Comparing the Clinical Utility and Diagnostic Performance of CSF P-Tau181, P-Tau217, and P-Tau231 Assays. Neurology 2021; 97:e1681-e1694. [PMID: 34493616 PMCID: PMC8605616 DOI: 10.1212/wnl.0000000000012727] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 08/19/2021] [Indexed: 12/30/2022] Open
Abstract
Background and Objectives Phosphorylated tau (p-tau) in CSF is considered an important biomarker in Alzheimer disease (AD) and has been incorporated in recent diagnostic criteria. Several variants exist, including p-tau at threonines 181 (p-tau181), 217 (p-tau217), and 231 (p-tau231). However, no studies have compared their diagnostic performance or association to β-amyloid (Aβ) and tau-PET. Understanding which p-tau variant to use remains an important yet answered question. We aimed to compare the diagnostic accuracy of p-tau181, p-tau217, and p-tau231 in CSF for AD and their association with Aβ and tau-PET. Methods A total of 629 participants in the Swedish BioFINDER-2 study were included (cognitively unimpaired, n = 334; Aβ-positive mild cognitive impairment, n = 84; AD dementia, n = 119; and non-AD disorders, n = 92). In addition to p-tau181 and p-tau217 measured using assays with the same detector antibodies from Eli Lilly (p-tau181Lilly, p-tau217Lilly) and p-tau231, we also included p-tau181 measurements from 2 commonly used assays (Innotest and Elecsys). Results Although all p-tau variants increased across the AD continuum, p-tau217Lilly showed the greatest dynamic range (13-fold increase vs 1.9–5.4-fold increase for other p-tau variants for AD dementia vs non-AD). P-Tau217Lilly showed stronger correlations with Aβ- and tau-PET (p < 0.0001). P-Tau217Lilly exhibited higher accuracy than other p-tau variants for separating AD dementia from non-AD (area under the curve [AUC], 0.98 vs 0.88 [p < 0.0001] - 0.96 [p < 0.05]) and for identifying Aβ-PET (AUC, 0.86 vs 0.74 [p < 0.0001] and 0.83 [p < 0.001]) and tau-PET positivity (AUC, 0.94 vs 0.80—0.92, p < 0.0001). Finally, p-Tau181Lilly generally performed better than the other p-tau181 assays (e.g., AD dementia vs non-AD, AUC, 0.96 vs 0.88 [p-tau181Innotest] and 0.89 [p-tau181Elecsys]; p < 0.0001). Discussion CSF p-tau217Lilly seems to be more useful than other included p-tau assays in the workup of AD. Varied results across p-tau181 assays highlights the importance of anti-tau antibodies for biomarker performance. Classification of Evidence This study provides Class II evidence that p-tau217 provides higher diagnostic accuracy for diagnosis of AD dementia than p-tau181 or p-tau231.
Collapse
Affiliation(s)
- Antoine Leuzy
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN.
| | - Shorena Janelidze
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Dirk Jacobs
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Claudia Cicognola
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Erik Stomrud
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Eugeen Vanmechelen
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Jeffrey L Dage
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN
| | - Oskar Hansson
- From the Clinical Memory Research Unit (A.L., S.J., N.M.-C., S.P., C.C., E.S., O.H.), Department of Clinical Sciences, Lund University, Malmö; Department of Neurology (N.M.-C.) and Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Lund; Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden; ADx NeuroSciences NV (D.J., E.V.), Ghent, Belgium; and Eli Lilly and Company (J.L.D.), Indianapolis, IN.
| |
Collapse
|
156
|
Carroll T, Guha S, Nehrke K, Johnson GVW. Tau Post-Translational Modifications: Potentiators of Selective Vulnerability in Sporadic Alzheimer's Disease. BIOLOGY 2021; 10:1047. [PMID: 34681146 PMCID: PMC8533264 DOI: 10.3390/biology10101047] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022]
Abstract
Sporadic Alzheimer's Disease (AD) is the most common form of dementia, and its severity is characterized by the progressive formation of tau neurofibrillary tangles along a well-described path through the brain. This spatial progression provides the basis for Braak staging of the pathological progression for AD. Tau protein is a necessary component of AD pathology, and recent studies have found that soluble tau species with selectively, but not extensively, modified epitopes accumulate along the path of disease progression before AD-associated insoluble aggregates form. As such, modified tau may represent a key cellular stressing agent that potentiates selective vulnerability in susceptible neurons during AD progression. Specifically, studies have found that tau phosphorylated at sites such as T181, T231, and S396 may initiate early pathological changes in tau by disrupting proper tau localization, initiating tau oligomerization, and facilitating tau accumulation and extracellular export. Thus, this review elucidates potential mechanisms through which tau post-translational modifications (PTMs) may simultaneously serve as key modulators of the spatial progression observed in AD development and as key instigators of early pathology related to neurodegeneration-relevant cellular dysfunctions.
Collapse
Affiliation(s)
- Trae Carroll
- Department of Pathology, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Sanjib Guha
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| |
Collapse
|
157
|
O'Connor A, Karikari TK, Poole T, Ashton NJ, Lantero Rodriguez J, Khatun A, Swift I, Heslegrave AJ, Abel E, Chung E, Weston PSJ, Pavisic IM, Ryan NS, Barker S, Rossor MN, Polke JM, Frost C, Mead S, Blennow K, Zetterberg H, Fox NC. Plasma phospho-tau181 in presymptomatic and symptomatic familial Alzheimer's disease: a longitudinal cohort study. Mol Psychiatry 2021; 26:5967-5976. [PMID: 32665603 PMCID: PMC7612227 DOI: 10.1038/s41380-020-0838-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 01/05/2023]
Abstract
Blood biomarkers have great potential to advance clinical care and accelerate trials in Alzheimer's disease (AD). Plasma phospho-tau181 (p-tau181) is a promising blood biomarker however, it is unknown if levels increase in presymptomatic AD. Therefore, we investigated the timing of p-tau181 changes using 153 blood samples from 70 individuals in a longitudinal study of familial AD (FAD). Plasma p-tau181 was measured, using an in-house single molecule array assay. We compared p-tau181 between symptomatic carriers, presymptomatic carriers, and non-carriers, adjusting for age and sex. We examined the relationship between p-tau181 and neurofilament light and estimated years to/from symptom onset (EYO), as well as years to/from actual onset in a symptomatic subgroup. In addition, we studied associations between p-tau181 and clinical severity, as well testing for differences between genetic subgroups. Twenty-four were presymptomatic carriers (mean baseline EYO -9.6 years) while 27 were non-carriers. Compared with non-carriers, plasma p-tau181 concentration was higher in both symptomatic (p < 0.001) and presymptomatic mutation carriers (p < 0.001). Plasma p-tau181 showed considerable intra-individual variability but individual values discriminated symptomatic (AUC 0.93 [95% CI 0.85-0.98]) and presymptomatic (EYO ≥ -7 years) (AUC 0.86 [95% CI 0.72-0.94]) carriers from non-carriers of the same age and sex. From a fitted model there was evidence (p = 0.050) that p-tau181 concentrations were higher in mutation carriers than non-carriers from 16 years prior to estimated symptom onset. Our finding that plasma p-tau181 concentration is increased in symptomatic and presymptomatic FAD suggests potential utility as an easily accessible biomarker of AD pathology.
Collapse
Affiliation(s)
- Antoinette O'Connor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Teresa Poole
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia, South London & Maudsley NHS Foundation, London, UK
| | - Juan Lantero Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Ayesha Khatun
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Imogen Swift
- UK Dementia Research Institute at UCL, London, UK
| | | | - Emily Abel
- UK Dementia Research Institute at UCL, London, UK
| | - Elisha Chung
- UK Dementia Research Institute at UCL, London, UK
| | - Philip S J Weston
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Ivanna M Pavisic
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Suzie Barker
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Martin N Rossor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - James M Polke
- Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Chris Frost
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Simon Mead
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London, UK.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK.
- UK Dementia Research Institute at UCL, London, UK.
| |
Collapse
|
158
|
Amro Z, Yool AJ, Collins-Praino LE. The potential role of glial cells in driving the prion-like transcellular propagation of tau in tauopathies. Brain Behav Immun Health 2021; 14:100242. [PMID: 34589757 PMCID: PMC8474563 DOI: 10.1016/j.bbih.2021.100242] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
Dementia is one of the leading causes of death worldwide, with tauopathies, a class of diseases defined by pathology associated with the microtubule-enriched protein, tau, as the major contributor. Although tauopathies, such as Alzheimer's disease and Frontotemporal dementia, are common amongst the ageing population, current effective treatment options are scarce, primarily due to the incomplete understanding of disease pathogenesis. The mechanisms via which aggregated forms of tau are able to propagate from one anatomical area to another to cause disease spread and progression is yet unknown. The prion-like hypothesis of tau propagation proposes that tau can propagate along neighbouring anatomical areas in a similar manner to prion proteins in prion diseases, such as Creutzfeldt-Jacob disease. This hypothesis has been supported by a plethora of studies that note the ability of tau to be actively secreted by neurons, propagated and internalised by neighbouring neuronal cells, causing disease spread. Surfacing research suggests a role of reactive astrocytes and microglia in early pre-clinical stages of tauopathy through their inflammatory actions. Furthermore, both glial types are able to internalise and secrete tau from the extracellular space, suggesting a potential role in tau propagation; although understanding the physiological mechanisms by which this can occur remains poorly understood. This review will discuss the current literature around the prion-like propagation of tau, with particular emphasis on glial-mediated neuroinflammation and the contribution it may play in this propagation process.
Collapse
Affiliation(s)
- Zein Amro
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | | |
Collapse
|
159
|
Xu Y, Du S, Marsh JA, Horie K, Sato C, Ballabio A, Karch CM, Holtzman DM, Zheng H. TFEB regulates lysosomal exocytosis of tau and its loss of function exacerbates tau pathology and spreading. Mol Psychiatry 2021; 26:5925-5939. [PMID: 32366951 PMCID: PMC7609570 DOI: 10.1038/s41380-020-0738-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 04/01/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
Neurofibrillary tangles (NFTs) composed of hyperphosphorylated and misfolded tau protein are a pathological hallmark of Alzheimer's disease and other tauopathy conditions. Tau is predominantly an intraneuronal protein but is also secreted in physiological and pathological conditions. The extracellular tau has been implicated in the seeding and propagation of tau pathology and is the prime target of the current tau immunotherapy. However, truncated tau species lacking the microtubule-binding repeat (MTBR) domains essential for seeding have been shown to undergo active secretion and the mechanisms and functional consequences of the various extracellular tau are poorly understood. We report here that the transcription factor EB (TFEB), a master regulator of lysosomal biogenesis, plays an essential role in the lysosomal exocytosis of selected tau species. TFEB loss of function significantly reduced the levels of interstitial fluid (ISF) tau in PS19 mice expressing P301S mutant tau and in conditioned media of mutant tau expressing primary neurons, while the secretion of endogenous wild-type tau was not affected. Mechanistically we found that TFEB regulates the secretion of truncated mutant tau lacking MTBR and this process is dependent on the lysosomal calcium channel TRPML1. Consistent with the seeding-incompetent nature of the truncated tau and supporting the concept that TFEB-mediated lysosomal exocytosis promotes cellular clearance, we show that reduced ISF tau in the absence of TFEB is associated with enhanced intraneuronal pathology and accelerated spreading. Our results support the idea that TFEB-mediated tau exocytosis serves as a clearance mechanism to reduce intracellular tau under pathological conditions and that effective tau immunotherapy should devoid targeting these extracellular tau species.
Collapse
Affiliation(s)
- Yin Xu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
| | - Shuqi Du
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jacob A. Marsh
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Andrea Ballabio
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA,Dan and Jan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA,Telethon Institute of Genetics and Medicine (TIGEM) and Department of Translational Medical Sciences, Frederico II University, Naples, Italy
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA,Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA,Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
160
|
Stamelou M, Respondek G, Giagkou N, Whitwell JL, Kovacs GG, Höglinger GU. Evolving concepts in progressive supranuclear palsy and other 4-repeat tauopathies. Nat Rev Neurol 2021; 17:601-620. [PMID: 34426686 DOI: 10.1038/s41582-021-00541-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Tauopathies are classified according to whether tau deposits predominantly contain tau isoforms with three or four repeats of the microtubule-binding domain. Those in which four-repeat (4R) tau predominates are known as 4R-tauopathies, and include progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, globular glial tauopathies and conditions associated with specific MAPT mutations. In these diseases, 4R-tau deposits are found in various cell types and anatomical regions of the brain and the conditions share pathological, pathophysiological and clinical characteristics. Despite being considered 'prototype' tauopathies and, therefore, ideal for studying neuroprotective agents, 4R-tauopathies are still severe and untreatable diseases for which no validated biomarkers exist. However, advances in research have addressed the issues of phenotypic overlap, early clinical diagnosis, pathophysiology and identification of biomarkers, setting a road map towards development of treatments. New clinical criteria have been developed and large cohorts with early disease are being followed up in prospective studies. New clinical trial readouts are emerging and biomarker research is focused on molecular pathways that have been identified. Lessons learned from failed trials of neuroprotective drugs are being used to design new trials. In this Review, we present an overview of the latest research in 4R-tauopathies, with a focus on progressive supranuclear palsy, and discuss how current evidence dictates ongoing and future research goals.
Collapse
Affiliation(s)
- Maria Stamelou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece. .,European University of Cyprus, Nicosia, Cyprus. .,Philipps University, Marburg, Germany.
| | - Gesine Respondek
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Nikolaos Giagkou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece
| | | | - Gabor G Kovacs
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Toronto, Ontario, Canada.,Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Günter U Höglinger
- Department of Neurology, Hanover Medical School, Hanover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
161
|
Barini E, Plotzky G, Mordashova Y, Hoppe J, Rodriguez-Correa E, Julier S, LePrieult F, Mairhofer I, Mezler M, Biesinger S, Cik M, Meinhardt MW, Ercan-Herbst E, Ehrnhoefer DE, Striebinger A, Bodie K, Klein C, Gasparini L, Schlegel K. Tau in the brain interstitial fluid is fragmented and seeding-competent. Neurobiol Aging 2021; 109:64-77. [PMID: 34655982 DOI: 10.1016/j.neurobiolaging.2021.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 11/19/2022]
Abstract
In Alzheimer disease, Tau pathology is thought to propagate from cell to cell throughout interconnected brain areas. However, the forms of Tau released into the brain interstitial fluid (ISF) in vivo during the development of Tauopathy and their pathological relevance remain unclear. Combining in vivo microdialysis and biochemical analysis, we find that in Tau transgenic mice, human Tau (hTau) present in brain ISF is truncated and comprises at least 10 distinct fragments spanning the entire Tau protein. The fragmentation pattern is similar across different Tau transgenic models, pathological stages and brain areas. ISF hTau concentration decreases during Tauopathy progression, while its phosphorylation increases. ISF from mice with established Tauopathy induces Tau aggregation in HEK293-Tau biosensor cells. Notably, immunodepletion of ISF phosphorylated Tau, but not Tau fragments, significantly reduces its ability to seed Tau aggregation and only a fraction of Tau, separated by ultracentrifugation, is seeding-competent. These results indicate that ISF seeding competence is driven by a small subset of Tau, which potentially contribute to the propagation of Tau pathology.
Collapse
Affiliation(s)
- Erica Barini
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany.
| | - Gudrun Plotzky
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Yulia Mordashova
- AbbVie Deutschland GmbH & Co. KG, Discovery and Exploratory Statistics (DIVES), Knollstrasse, Ludwigshafen, Germany
| | - Jonas Hoppe
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Esther Rodriguez-Correa
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Sonja Julier
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Florie LePrieult
- AbbVie Deutschland GmbH & Co. KG, Drug Metabolism and Pharmacokinetics, Knollstrasse, Ludwigshafen, Germany
| | - Ina Mairhofer
- AbbVie Deutschland GmbH & Co. KG, Drug Metabolism and Pharmacokinetics, Knollstrasse, Ludwigshafen, Germany
| | - Mario Mezler
- AbbVie Deutschland GmbH & Co. KG, Drug Metabolism and Pharmacokinetics, Knollstrasse, Ludwigshafen, Germany
| | - Sandra Biesinger
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Miroslav Cik
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Marcus W Meinhardt
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | | | - Dagmar E Ehrnhoefer
- BioMed X GmbH, Im Neuenheimer Feld, Heidelberg, Germany; AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Andreas Striebinger
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Karen Bodie
- AbbVie Deutschland GmbH & Co. KG, Preclinical Safety, Knollstrasse, Ludwigshafen, Germany
| | - Corinna Klein
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany
| | - Laura Gasparini
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany.
| | - Kerstin Schlegel
- AbbVie Deutschland GmbH & Co. KG , Neuroscience Discovery, Knollstrasse, Ludwigshafen, Germany.
| |
Collapse
|
162
|
Hier DB, Obafemi-Ajayi T, Thimgan MS, Olbricht GR, Azizi S, Allen B, Hadi BA, Wunsch DC. Blood biomarkers for mild traumatic brain injury: a selective review of unresolved issues. Biomark Res 2021; 9:70. [PMID: 34530937 PMCID: PMC8447604 DOI: 10.1186/s40364-021-00325-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/26/2021] [Indexed: 01/03/2023] Open
Abstract
Background The use of blood biomarkers after mild traumatic brain injury (mTBI) has been widely studied. We have identified eight unresolved issues related to the use of five commonly investigated blood biomarkers: neurofilament light chain, ubiquitin carboxy-terminal hydrolase-L1, tau, S100B, and glial acidic fibrillary protein. We conducted a focused literature review of unresolved issues in three areas: mode of entry into and exit from the blood, kinetics of blood biomarkers in the blood, and predictive capacity of the blood biomarkers after mTBI. Findings Although a disruption of the blood brain barrier has been demonstrated in mild and severe traumatic brain injury, biomarkers can enter the blood through pathways that do not require a breach in this barrier. A definitive accounting for the pathways that biomarkers follow from the brain to the blood after mTBI has not been performed. Although preliminary investigations of blood biomarkers kinetics after TBI are available, our current knowledge is incomplete and definitive studies are needed. Optimal sampling times for biomarkers after mTBI have not been established. Kinetic models of blood biomarkers can be informative, but more precise estimates of kinetic parameters are needed. Confounding factors for blood biomarker levels have been identified, but corrections for these factors are not routinely made. Little evidence has emerged to date to suggest that blood biomarker levels correlate with clinical measures of mTBI severity. The significance of elevated biomarker levels thirty or more days following mTBI is uncertain. Blood biomarkers have shown a modest but not definitive ability to distinguish concussed from non-concussed subjects, to detect sub-concussive hits to the head, and to predict recovery from mTBI. Blood biomarkers have performed best at distinguishing CT scan positive from CT scan negative subjects after mTBI.
Collapse
Affiliation(s)
- Daniel B Hier
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.
| | - Tayo Obafemi-Ajayi
- Cooperative Engineering Program, Missouri State University, Springfield, MO 65897, United States
| | - Matthew S Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Sima Azizi
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Blaine Allen
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Bassam A Hadi
- Department of Surgery, Mercy Hospital, St. Louis MO, Missouri, MO 63141, United States
| | - Donald C Wunsch
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.,National Science Foundation, ECCS Division, Virginia, 22314, USA
| |
Collapse
|
163
|
Van Kolen K, Malia TJ, Theunis C, Nanjunda R, Teplyakov A, Ernst R, Wu SJ, Luo J, Borgers M, Vandermeeren M, Bottelbergs A, Wintmolders C, Lacy E, Maurin H, Larsen P, Willems R, Van De Casteele T, Triana-Baltzer G, Slemmon R, Galpern W, Trojanowski JQ, Sun H, Mercken MH. Discovery and Functional Characterization of hPT3, a Humanized Anti-Phospho Tau Selective Monoclonal Antibody. J Alzheimers Dis 2021; 77:1397-1416. [PMID: 32894244 DOI: 10.3233/jad-200544] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND As a consequence of the discovery of an extracellular component responsible for the progression of tau pathology, tau immunotherapy is being extensively explored in both preclinical and clinical studies as a disease modifying strategy for the treatment of Alzheimer's disease. OBJECTIVE Describe the characteristics of the anti-phospho (T212/T217) tau selective antibody PT3 and its humanized variant hPT3. METHODS By performing different immunization campaigns, a large collection of antibodies has been generated and prioritized. In depth, in vitro characterization using surface plasmon resonance, phospho-epitope mapping, and X-ray crystallography experiments were performed. Further characterization involved immunohistochemical staining on mouse- and human postmortem tissue and neutralization of tau seeding by immunodepletion assays. RESULTS AND CONCLUSION Various in vitro experiments demonstrated a high intrinsic affinity for PT3 and hPT3 for AD brain-derived paired helical filaments but also to non-aggregated phospho (T212/T217) tau. Further functional analyses in cellular and in vivo models of tau seeding demonstrated almost complete depletion of tau seeds in an AD brain homogenate. Ongoing trials will provide the clinical evaluation of the tau spreading hypothesis in Alzheimer's disease.
Collapse
Affiliation(s)
- Kristof Van Kolen
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Thomas J Malia
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Clara Theunis
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Rupesh Nanjunda
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Alexey Teplyakov
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Robin Ernst
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Sheng-Jiun Wu
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Jinquan Luo
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Marianne Borgers
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Marc Vandermeeren
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Astrid Bottelbergs
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Cindy Wintmolders
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Eilyn Lacy
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Hervé Maurin
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Peter Larsen
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Roland Willems
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Tom Van De Casteele
- Translational Medicine and Early Development Statistics Janssen Research & Development, Beerse, Belgium
| | | | - Randy Slemmon
- Neuroscience biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Wendy Galpern
- Neuroscience Experimental medicine, Janssen Research & Development, Titusville, NJ, USA
| | | | - Hong Sun
- Neuroscience Clinical Development, Janssen Research & Development, Titusville, NJ, USA
| | - Marc H Mercken
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| |
Collapse
|
164
|
Triana-Baltzer G, Van Kolen K, Theunis C, Moughadam S, Slemmon R, Mercken M, Galpern W, Sun H, Kolb H. Development and Validation of a High Sensitivity Assay for Measuring p217 + tau in Cerebrospinal Fluid. J Alzheimers Dis 2021; 77:1417-1430. [PMID: 32831201 PMCID: PMC7683057 DOI: 10.3233/jad-200463] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Early and accurate detection and staging is critical to managing Alzheimer’s disease (AD) and supporting clinical trials. Cerebrospinal fluid (CSF) biomarkers for amyloid-β peptides, tau species, and various neurodegenerative and inflammatory analytes are leading the way in this regard, yet there is room for improved sensitivity and specificity. In particular tau is known to be present in many different fragments, conformations, and post-translationally modified forms. While the exact tau species that might best reflect AD pathology is unknown, a growing body of evidence suggests that forms with high levels of phosphorylation in the mid-region may be especially enriched in AD. Objective: Develop an assay for measuring p217tau in CSF. Methods: Here we describe the development and validation of a novel sELISA for measuring CSF tau species containing phosphorylation at threonines 212 & 217, aka p217 + tau, using the PT3 antibody. Results: While the analyte is present at extremely low levels the assay is sufficiently sensitive and specific to quantitate p217 + tau with excellent precision, accuracy, and dilution linearity, allowing good differentiation between diagnostic subgroups. The p217 + tau measurements appear to track AD pathology better than the commonly used p181tau epitope, suggesting superior diagnostic and staging performance. Finally, the assay can also be configured to differentiate antibody-bound versus antibody-free tau, and therefore can be used to measure target engagement by p217 + tau-targeting immunotherapeutics. Conclusion: The assay for measuring p217 + tau described here is highly sensitive, accurate, precise, dilution linear, and shows good potential for identifying and staging AD.
Collapse
Affiliation(s)
| | - Kristof Van Kolen
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Clara Theunis
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Setareh Moughadam
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Randy Slemmon
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Marc Mercken
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Wendy Galpern
- Neuroscience Experimental Medicine, Janssen Research & Development, Titusville, NJ, USA
| | - Hong Sun
- Neuroscience Clinical Development, Janssen Research & Development, Titusville, NJ, USA
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, CA, USA
| |
Collapse
|
165
|
Vogels T, Vargová G, Brezováková V, Quint WH, Hromádka T. Viral Delivery of Non-Mutated Human Truncated Tau to Neurons Recapitulates Key Features of Human Tauopathy in Wild-Type Mice. J Alzheimers Dis 2021; 77:551-568. [PMID: 32675411 DOI: 10.3233/jad-200047] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Neuronal accumulation of hyperphosphorylated and truncated tau aggregates is one of the major defining factors and key drivers of neurodegeneration in Alzheimer's disease and other tauopathies. OBJECTIVE We developed an AAV-induced model of tauopathy mediated by human truncated tau protein without familial frontotemporal dementia-related mutations to study tau propagation and the functional consequences of tau pathology. METHODS We performed targeted transductions of the hippocampus or entorhinal cortex in adult mice followed by histological analysis to study the progression of hippocampal tau pathology and tau spreading. We performed behavioral analysis of mice with AAV-induced hippocampal tau pathology. RESULTS AAV-induced hippocampal tau pathology was characterized by tau hyperphosphorylation (AT8 positivity), sarkosyl insolubility, and the presence of neurofibrillary tangles. AAV-induced tau pathology was associated with microgliosis and hypertrophic astrocytes in the absence of cognitive deficits. Additionally, the co-expression of mCherry fluorescent protein and human truncated tau enabled us to detect both local spreading of human tau and spreading from the entorhinal cortex to the synaptically connected dentate gyrus. CONCLUSION Targeted delivery of AAV with truncated tau protein into subcortical and cortical structures of mammalian brains represents an efficient approach for creating temporally and spatially well-defined tau pathology suitable for in vivo studies of tau propagation and neuronal circuit deficits in Alzheimer's disease.
Collapse
Affiliation(s)
- Thomas Vogels
- Axon Neuroscience R & D Services SE, Bratislava, Slovakia
| | - Gréta Vargová
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | - Tomáš Hromádka
- Axon Neuroscience R & D Services SE, Bratislava, Slovakia.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
166
|
Halaas NB, Zetterberg H, Idland AV, Knapskog AB, Watne LO, Blennow K. Cerebrospinal Fluid Concentration of Neurogranin in Hip Fracture Patients with Delirium. J Alzheimers Dis 2021; 81:667-677. [PMID: 33814433 DOI: 10.3233/jad-201341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Delirium is associated with an increased risk of incident dementia and accelerated progression of existing cognitive symptoms. Reciprocally, dementia increases the risk of delirium. Cerebrospinal fluid (CSF) concentration of the dendritic protein neurogranin has been shown to increase in early Alzheimer's disease (AD), likely reflecting synaptic dysfunction and/or degeneration. OBJECTIVE To elucidate the involvement of synaptic dysfunction in delirium pathophysiology, we tested the association between CSF neurogranin concentration and delirium in hip fracture patients with different AD-biomarker profiles, while comparing them to cognitively unimpaired older adults (CUA) and AD patients. METHODS The cohort included hip fracture patients with (n = 70) and without delirium (n = 58), CUA undergoing elective surgery (n = 127), and AD patients (n = 46). CSF was collected preoperatively and diagnostically in surgery and AD patients respectively. CSF neurogranin concentrations were analyzed in all samples with an in-house ELISA. Delirium was assessed pre-and postoperatively in hip fracture patients by trained investigators using the Confusion Assessment Method. Hip fracture patients were further stratified based on pre-fracture dementia status, delirium subtype, and AD fluid biomarkers. RESULTS No association was found between delirium and CSF neurogranin concentration (main analysis: delirium versus no delirium, p = 0.68). Hip fracture patients had lower CSF neurogranin concentration than AD patients (p = 0.001) and CUA (p = 0.035) in age-adjusted sensitivity analyses. CONCLUSION The findings suggest that delirium is not associated with increased CSF neurogranin concentration in hip fracture patients, possibly due to advanced neurodegenerative disease and age and/or because synaptic degeneration is not an important pathophysiological process in delirium.
Collapse
Affiliation(s)
- Nathalie Bodd Halaas
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.,Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom.,UK Dementia Research Institute at UCL, London, United Kingdom
| | - Ane-Victoria Idland
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Leiv Otto Watne
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
167
|
McGuigan S, Evered L, Scott DA, Silbert B, Zetterberg H, Blennow K. Comparing the effect of xenon and sevoflurane anesthesia on postoperative neural injury biomarkers: a randomized controlled trial. Med Gas Res 2021; 12:10-17. [PMID: 34472497 PMCID: PMC8447955 DOI: 10.4103/2045-9912.324591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
General anesthesia and surgery are associated with an increase in neural injury biomarkers. Elevations of these neural injury biomarkers in the perioperative period are associated with postoperative delirium. Xenon has been shown to be protective against a range of neurological insults in animal models. It remains to be seen if xenon anesthesia is neuroprotective in the perioperative setting in humans. Twenty-four participants scheduled for lithotripsy were randomized to receive either xenon or sevoflurane general anesthesia. There was no statistically significant difference in the concentrations of postoperative neural injury biomarkers between the xenon and sevoflurane group. Following the procedure there was a significant increase in the concentration from baseline of all three biomarkers at 1 hour post-induction with a return to baseline at 5 hours. General anesthesia for lithotripsy was associated with a significant increase at 1 hour post-induction in the neural injury biomarkers total tau, neurofilament light and tau phosphorylated at threonine 181, a marker of tau phosphorylation. The protocol was approved by the St. Vincent’s Hospital Melbourne Ethics Committee (approval No. HREC/18/SVHM/221) on July 20, 2018 and was registered with the Australia New Zealand Clinical Trials Registry (registration No. ACTRN12618000916246) on May 31, 2018.
Collapse
Affiliation(s)
- Steven McGuigan
- Department of Anaesthesia and Acute Pain Medicine, St. Vincent's Hospital; Department of Critical Care, University of Melbourne, Melbourne, Australia
| | - Lisbeth Evered
- Department of Anaesthesia and Acute Pain Medicine, St. Vincent's Hospital; Department of Critical Care, University of Melbourne, Melbourne, Australia; Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - David A Scott
- Department of Anaesthesia and Acute Pain Medicine, St. Vincent's Hospital; Department of Critical Care, University of Melbourne, Melbourne, Australia
| | - Brendan Silbert
- Department of Anaesthesia and Acute Pain Medicine, St. Vincent's Hospital; Department of Critical Care, University of Melbourne, Melbourne, Australia
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology; UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
168
|
Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simrén J, Montoliu‐Gaya L, Pannee J, O´Connor A, Weston PSJ, Lantero‐Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, Zetterberg H. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease. J Intern Med 2021; 290:583-601. [PMID: 34021943 PMCID: PMC8416781 DOI: 10.1111/joim.13332] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is increasingly prevalent worldwide, and disease-modifying treatments may soon be at hand; hence, now, more than ever, there is a need to develop techniques that allow earlier and more secure diagnosis. Current biomarker-based guidelines for AD diagnosis, which have replaced the historical symptom-based guidelines, rely heavily on neuroimaging and cerebrospinal fluid (CSF) sampling. While these have greatly improved the diagnostic accuracy of AD pathophysiology, they are less practical for application in primary care, population-based and epidemiological settings, or where resources are limited. In contrast, blood is a more accessible and cost-effective source of biomarkers in AD. In this review paper, using the recently proposed amyloid, tau and neurodegeneration [AT(N)] criteria as a framework towards a biological definition of AD, we discuss recent advances in biofluid-based biomarkers, with a particular emphasis on those with potential to be translated into blood-based biomarkers. We provide an overview of the research conducted both in CSF and in blood to draw conclusions on biomarkers that show promise. Given the evidence collated in this review, plasma neurofilament light chain (N) and phosphorylated tau (p-tau; T) show particular potential for translation into clinical practice. However, p-tau requires more comparisons to be conducted between its various epitopes before conclusions can be made as to which one most robustly differentiates AD from non-AD dementias. Plasma amyloid beta (A) would prove invaluable as an early screening modality, but it requires very precise tests and robust pre-analytical protocols.
Collapse
Affiliation(s)
- D. O. T. Alawode
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - A. J. Heslegrave
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - N. J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing’s College LondonLondonUK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS FoundationLondonUK
| | - T. K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Simrén
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - L. Montoliu‐Gaya
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Pannee
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - A. O´Connor
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - P. S. J. Weston
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - A. Keshavan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - A. Snellman
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Turku PET CentreUniversity of TurkuTurkuFinland
| | - J. Gobom
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - R. W. Paterson
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. M. Schott
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - K. Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - N. C. Fox
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - H. Zetterberg
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| |
Collapse
|
169
|
Moloney CM, Lowe VJ, Murray ME. Visualization of neurofibrillary tangle maturity in Alzheimer's disease: A clinicopathologic perspective for biomarker research. Alzheimers Dement 2021; 17:1554-1574. [PMID: 33797838 PMCID: PMC8478697 DOI: 10.1002/alz.12321] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/11/2021] [Accepted: 02/03/2021] [Indexed: 12/29/2022]
Abstract
Neurofibrillary tangles, one of the neuropathologic hallmarks of Alzheimer's disease, have a dynamic lifespan of maturity that associates with progressive neuronal dysfunction and cognitive deficits. As neurofibrillary tangles mature, the biology of the neuron undergoes extensive changes that may impact biomarker recognition and therapeutic targeting. Neurofibrillary tangle maturity encompasses three levels: pretangles, mature tangles, and ghost tangles. In this review, we detail distinct and overlapping characteristics observed in the human brain regarding morphologic changes the neuron undergoes, conversion from intracellular to extracellular space, tau immunostaining patterns, and tau isoform expression changes across the lifespan of the neurofibrillary tangle. Post-translational modifications of tau such as phosphorylation, ubiquitination, conformational events, and truncations are discussed to contextualize tau immunostaining patterns. We summarize accumulated and emerging knowledge of neurofibrillary tangle maturity, discuss the current tools used to interpret the dynamic nature in the postmortem brain, and consider implications for cognitive dysfunction and tau biomarkers.
Collapse
Affiliation(s)
| | - Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | | |
Collapse
|
170
|
Sinsky J, Pichlerova K, Hanes J. Tau Protein Interaction Partners and Their Roles in Alzheimer's Disease and Other Tauopathies. Int J Mol Sci 2021; 22:9207. [PMID: 34502116 PMCID: PMC8431036 DOI: 10.3390/ijms22179207] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Tau protein plays a critical role in the assembly, stabilization, and modulation of microtubules, which are important for the normal function of neurons and the brain. In diseased conditions, several pathological modifications of tau protein manifest. These changes lead to tau protein aggregation and the formation of paired helical filaments (PHF) and neurofibrillary tangles (NFT), which are common hallmarks of Alzheimer's disease and other tauopathies. The accumulation of PHFs and NFTs results in impairment of physiological functions, apoptosis, and neuronal loss, which is reflected as cognitive impairment, and in the late stages of the disease, leads to death. The causes of this pathological transformation of tau protein haven't been fully understood yet. In both physiological and pathological conditions, tau interacts with several proteins which maintain their proper function or can participate in their pathological modifications. Interaction partners of tau protein and associated molecular pathways can either initiate and drive the tau pathology or can act neuroprotective, by reducing pathological tau proteins or inflammation. In this review, we focus on the tau as a multifunctional protein and its known interacting partners active in regulations of different processes and the roles of these proteins in Alzheimer's disease and tauopathies.
Collapse
Affiliation(s)
| | | | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia; (J.S.); (K.P.)
| |
Collapse
|
171
|
Brain injury markers in new-onset seizures in adults: A pilot study. Seizure 2021; 92:62-67. [PMID: 34455195 DOI: 10.1016/j.seizure.2021.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/07/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Biochemical markers of brain pathology could potentially contribute to diagnosis and prediction in epilepsy. We describe levels of five brain injury markers in adults with new-onset seizures, and assess group differences in patients with a single seizure, epilepsy, and poststroke epilepsy. METHODS In this prospective observational study, adults with new-onset seizures were recruited at Sahlgrenska University Hospital, Sweden, and concentrations of glial fibrillary acidic protein (GFAP), neurofilament light (NfL), microtubule-associated protein tau (tau), S100 calcium-binding protein (S100B), and neuron-specific enolase (NSE) were measured. Participants were categorized as epilepsy, poststroke epilepsy (PSE), or single seizure (no additional seizures). Patients were followed until a diagnosis of epilepsy or PSE, or for at least two years in single seizure cases. RESULTS The cohort included 23 (37%) individuals with a single seizure, 24 (39%) with epilepsy, and 15 (24%) with PSE. The concentrations of S100B were higher in patients with epilepsy and PSE than in single seizures (p = 0.0023 and p = 0.0162, respectively). The concentrations of NfL were higher in patients with PSE than in single seizures (p=0.0027). After age-normalization, levels of S100B were higher in patients with epilepsy and levels of NfL were higher in patients with PSE (p = 0.0021 and p = 0.0180). CONCLUSION Levels of S100B and NfL were higher in patients with epilepsy or PSE than patients with single seizures. Further studies are needed to investigate the biomarker potential of brain injury markers as predictors of epilepsy course or indicators of epileptogenesis.
Collapse
|
172
|
Bowles KR, Silva MC, Whitney K, Bertucci T, Berlind JE, Lai JD, Garza JC, Boles NC, Mahali S, Strang KH, Marsh JA, Chen C, Pugh DA, Liu Y, Gordon RE, Goderie SK, Chowdhury R, Lotz S, Lane K, Crary JF, Haggarty SJ, Karch CM, Ichida JK, Goate AM, Temple S. ELAVL4, splicing, and glutamatergic dysfunction precede neuron loss in MAPT mutation cerebral organoids. Cell 2021; 184:4547-4563.e17. [PMID: 34314701 PMCID: PMC8635409 DOI: 10.1016/j.cell.2021.07.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/06/2021] [Accepted: 06/30/2021] [Indexed: 12/21/2022]
Abstract
Frontotemporal dementia (FTD) because of MAPT mutation causes pathological accumulation of tau and glutamatergic cortical neuronal death by unknown mechanisms. We used human induced pluripotent stem cell (iPSC)-derived cerebral organoids expressing tau-V337M and isogenic corrected controls to discover early alterations because of the mutation that precede neurodegeneration. At 2 months, mutant organoids show upregulated expression of MAPT, glutamatergic signaling pathways, and regulators, including the RNA-binding protein ELAVL4, and increased stress granules. Over the following 4 months, mutant organoids accumulate splicing changes, disruption of autophagy function, and build-up of tau and P-tau-S396. By 6 months, tau-V337M organoids show specific loss of glutamatergic neurons as seen in individuals with FTD. Mutant neurons are susceptible to glutamate toxicity, which can be rescued pharmacologically by the PIKFYVE kinase inhibitor apilimod. Our results demonstrate a sequence of events that precede neurodegeneration, revealing molecular pathways associated with glutamate signaling as potential targets for therapeutic intervention in FTD.
Collapse
Affiliation(s)
- Kathryn R Bowles
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - M Catarina Silva
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kristen Whitney
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | | | - Joshua E Berlind
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jesse D Lai
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Amgen Research, One Amgen Center Dr., Thousand Oaks, CA 91320, USA
| | - Jacob C Garza
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Sidhartha Mahali
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kevin H Strang
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | - Jacob A Marsh
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Cynthia Chen
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Derian A Pugh
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Yiyuan Liu
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Ronald E Gordon
- Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | | | | | - Steven Lotz
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Keith Lane
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - John F Crary
- Department of Pathology, Neuropathology Brain Bank and Research Core, ISMMS, New York, NY 10029, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Departments of Genetics and Genomic Sciences, Neuroscience, and Neurology, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA.
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
173
|
Prakash P, Jethava KP, Korte N, Izquierdo P, Favuzzi E, Rose IVL, Guttenplan KA, Manchanda P, Dutta S, Rochet JC, Fishell G, Liddelow SA, Attwell D, Chopra G. Monitoring phagocytic uptake of amyloid β into glial cell lysosomes in real time. Chem Sci 2021; 12:10901-10918. [PMID: 34476070 PMCID: PMC8372545 DOI: 10.1039/d1sc03486c] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/30/2022] Open
Abstract
Phagocytosis by glial cells is essential to regulate brain function during health and disease. Therapies for Alzheimer's disease (AD) have primarily focused on targeting antibodies to amyloid β (Aβ) or inhibitng enzymes that make it, and while removal of Aβ by phagocytosis is protective early in AD it remains poorly understood. Impaired phagocytic function of glial cells during later stages of AD likely contributes to worsened disease outcome, but the underlying mechanisms of how this occurs remain unknown. We have developed a human Aβ1-42 analogue (AβpH) that exhibits green fluorescence upon internalization into the acidic organelles of cells but is non-fluorescent at physiological pH. This allowed us to image, for the first time, glial uptake of AβpH in real time in live animals. We find that microglia phagocytose more AβpH than astrocytes in culture, in brain slices and in vivo. AβpH can be used to investigate the phagocytic mechanisms responsible for removing Aβ from the extracellular space, and thus could become a useful tool to study Aβ clearance at different stages of AD.
Collapse
Affiliation(s)
- Priya Prakash
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
| | - Krupal P Jethava
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
| | - Nils Korte
- Department of Neuroscience, Physiology and Pharmacology, University College London London WC1E 6BT UK
| | - Pablo Izquierdo
- Department of Neuroscience, Physiology and Pharmacology, University College London London WC1E 6BT UK
| | - Emilia Favuzzi
- Department of Neurobiology, Harvard Medical School 220 Longwood Avenue Boston MA 02115 USA
- Stanley Center at the Broad 75 Ames Street Cambridge MA 02142 USA
| | - Indigo V L Rose
- Neuroscience Institute, NYU Grossman School of Medicine New York NY 10016 USA
| | | | - Palak Manchanda
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
| | - Sayan Dutta
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette IN 47907 USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette IN 47907 USA
- Purdue Institute for Integrative Neuroscience, Purdue University West Lafayette IN 47907 USA
| | - Gord Fishell
- Department of Neurobiology, Harvard Medical School 220 Longwood Avenue Boston MA 02115 USA
- Stanley Center at the Broad 75 Ames Street Cambridge MA 02142 USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine New York NY 10016 USA
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine New York NY 10016 USA
- Department of Ophthalmology, NYU Grossman School of Medicine New York NY 10016 USA
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London London WC1E 6BT UK
| | - Gaurav Chopra
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
- Purdue Institute for Integrative Neuroscience, Purdue University West Lafayette IN 47907 USA
- Purdue Institute for Drug Discovery 720 Clinic Drive West Lafayette IN 47907 USA
- Purdue Center for Cancer Research, Purdue University West Lafayette IN 47907 USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University West Lafayette IN 47907 USA
| |
Collapse
|
174
|
Genetic and environmental factors in Alzheimer's and Parkinson's diseases and promising therapeutic intervention via fecal microbiota transplantation. NPJ Parkinsons Dis 2021; 7:70. [PMID: 34381040 PMCID: PMC8357954 DOI: 10.1038/s41531-021-00213-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases are characterized by neuronal impairment and loss of function, and with the major shared histopathological hallmarks of misfolding and aggregation of specific proteins inside or outside cells. Some genetic and environmental factors contribute to the promotion of the development and progression of neurodegenerative diseases. Currently, there are no effective treatments for neurodegenerative diseases. It has been revealed that bidirectional communication exists between the brain and the gut. The gut microbiota is a changeable and experience-dependent ecosystem and can be modified by genetic and environmental factors. The gut microbiota provides potential therapeutic targets that can be regulated as new interventions for neurodegenerative diseases. In this review, we discuss genetic and environmental risk factors for neurodegenerative diseases, summarize the communication among the components of the microbiota-gut-brain axis, and discuss the treatment strategy of fecal microbiota transplantation (FMT). FMT is a promising treatment for neurodegenerative diseases, and restoration of the gut microbiota to a premorbid state is a novel goal for prevention and treatment strategies.
Collapse
|
175
|
Abstract
This scientific commentary refers to ‘CSF tau microtubule binding region identifies tau tangle and clinical stages of Alzheimer’s disease’, by Horie et al. (doi:10.1093/brain/awaa373).
Collapse
Affiliation(s)
- Jamie Toombs
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, The University of Edinburgh, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
176
|
Sato C, Mallipeddi N, Ghoshal N, Wright BA, Day GS, Davis AA, Kim AH, Zipfel GJ, Bateman RJ, Gabelle A, Barthélemy NR. MAPT R406W increases tau T217 phosphorylation in absence of amyloid pathology. Ann Clin Transl Neurol 2021; 8:1817-1830. [PMID: 34342183 PMCID: PMC8419397 DOI: 10.1002/acn3.51435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 01/07/2023] Open
Abstract
Objective Tau hyperphosphorylation at threonine 217 (pT217) in cerebrospinal fluid (CSF) has recently been linked to early amyloidosis and could serve as a highly sensitive biomarker for Alzheimer’s disease (AD). However, it remains unclear whether other tauopathies induce pT217 modifications. To determine if pT217 modification is specific to AD, CSF pT217 was measured in AD and other tauopathies. Methods Using immunoprecipitation and mass spectrometry methods, we compared CSF T217 phosphorylation occupancy (pT217/T217) and amyloid‐beta (Aβ) 42/40 ratio in cognitively normal individuals and those with symptomatic AD, progressive supranuclear palsy, corticobasal syndrome, and sporadic and familial frontotemporal dementia. Results Individuals with AD had high CSF pT217/T217 and low Aβ42/40. In contrast, cognitively normal individuals and the majority of those with 4R tauopathies had low CSF pT217/T217 and normal Aβ 42/40. We identified a subgroup of individuals with increased CSF pT217/T217 and normal Aβ 42/40 ratio, most of whom were MAPT R406W mutation carriers. Diagnostic accuracies of CSF Aβ 42/40 and CSF pT217/T217, alone and in combination were compared. We show that CSF pT217/T217 × CSF Aβ 42/40 is a sensitive composite biomarker that can separate MAPT R406W carriers from cognitively normal individuals and those with other tauopathies. Interpretation MAPT R406W is a tau mutation that leads to 3R+4R tauopathy similar to AD, but without amyloid neuropathology. These findings suggest that change in CSF pT217/T217 ratio is not specific to AD and might reflect common downstream tau pathophysiology common to 3R+4R tauopathies.
Collapse
Affiliation(s)
- Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Nipun Mallipeddi
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Nupur Ghoshal
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Brenton A Wright
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, California
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida
| | - Albert A Davis
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri
| | - Albert H Kim
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri.,Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gregory J Zipfel
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri.,Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri.,Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Audrey Gabelle
- Department of Neurology, Memory Research and Resources Center, University Hospital of Montpellier, Neurosciences Institute of Montpellier, University of Montpellier, Montpellier, France
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
177
|
Safety and efficacy of anti-tau monoclonal antibody gosuranemab in progressive supranuclear palsy: a phase 2, randomized, placebo-controlled trial. Nat Med 2021; 27:1451-1457. [PMID: 34385707 DOI: 10.1038/s41591-021-01455-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
A randomized, double-blind, placebo-controlled, 52-week study (no. NCT03068468) evaluated gosuranemab, an anti-tau monoclonal antibody, in the treatment of progressive supranuclear palsy (PSP). In total, 486 participants dosed were assigned to either gosuranemab (n = 321) or placebo (n = 165). Efficacy was not demonstrated on adjusted mean change of PSP Rating Scale score at week 52 between gosuranemab and placebo (10.4 versus 10.6, P = 0.85, primary endpoint), or at secondary endpoints, resulting in discontinuation of the open-label, long-term extension. Unbound N-terminal tau in cerebrospinal fluid decreased by 98% with gosuranemab and increased by 11% with placebo (P < 0.0001). Incidences of adverse events and deaths were similar between groups. This well-powered study suggests that N-terminal tau neutralization does not translate into clinical efficacy.
Collapse
|
178
|
Trudler D, Ghatak S, Lipton SA. Emerging hiPSC Models for Drug Discovery in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:8196. [PMID: 34360966 PMCID: PMC8347370 DOI: 10.3390/ijms22158196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide and are characterized by the chronic and progressive deterioration of neural function. Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), represent a huge social and economic burden due to increasing prevalence in our aging society, severity of symptoms, and lack of effective disease-modifying therapies. This lack of effective treatments is partly due to a lack of reliable models. Modeling neurodegenerative diseases is difficult because of poor access to human samples (restricted in general to postmortem tissue) and limited knowledge of disease mechanisms in a human context. Animal models play an instrumental role in understanding these diseases but fail to comprehensively represent the full extent of disease due to critical differences between humans and other mammals. The advent of human-induced pluripotent stem cell (hiPSC) technology presents an advantageous system that complements animal models of neurodegenerative diseases. Coupled with advances in gene-editing technologies, hiPSC-derived neural cells from patients and healthy donors now allow disease modeling using human samples that can be used for drug discovery.
Collapse
Affiliation(s)
- Dorit Trudler
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Swagata Ghatak
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Stuart A. Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
179
|
Shinohara M, Hirokawa J, Shimodaira A, Tashiro Y, Suzuki K, Gheni G, Fukumori A, Matsubara T, Morishima M, Saito Y, Murayama S, Sato N. ELISA Evaluation of Tau Accumulation in the Brains of Patients with Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:652-662. [PMID: 34283221 DOI: 10.1093/jnen/nlab047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite the routine use of sandwich enzyme-linked immunosorbent assays (ELISAs) for quantifying tau levels in CSF and plasma, tau accumulations in the brains of patients with Alzheimer disease (AD) have rarely been evaluated by this method. Thus, by introducing several tau ELISAs that target different epitopes, we evaluated accumulated tau levels in postmortem brains depending on disease stage, brain areas, and other AD-related changes. Notably, tau levels in insoluble fraction determined by each ELISAs differ depending on the epitopes of antibodies: non-AD control samples yield relatively high signals when an antibody against the N-terminal region of tau is used. On the other hand, ELISAs combining antibodies against the later-middle to C-terminal regions of tau produced substantially increased signals from AD samples, compared to those from non-AD controls. Such ELISAs better distinguish AD and non-AD controls, and the results are more closely associated with Braak neurofibrillary tangles stage, Aβ accumulation, and glial markers. Moreover, these ELISAs can reflect the pattern of tau spread across brain regions. In conclusion, Tau ELISAs that combine antibodies against the later-middle to C-terminal regions of tau can better reflect neuropathological tau accumulation, which would enable to evaluate tau accumulation in the brain at a biochemical level.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Junko Hirokawa
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Akemi Shimodaira
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Yoshitaka Tashiro
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Kaoru Suzuki
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Ghupurjan Gheni
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Akio Fukumori
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Tomoyasu Matsubara
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Maho Morishima
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Yuko Saito
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Shigeo Murayama
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Naoyuki Sato
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| |
Collapse
|
180
|
Ayalon G, Lee SH, Adolfsson O, Foo-Atkins C, Atwal JK, Blendstrup M, Booler H, Bravo J, Brendza R, Brunstein F, Chan R, Chandra P, Couch JA, Datwani A, Demeule B, DiCara D, Erickson R, Ernst JA, Foreman O, He D, Hötzel I, Keeley M, Kwok MCM, Lafrance-Vanasse J, Lin H, Lu Y, Luk W, Manser P, Muhs A, Ngu H, Pfeifer A, Pihlgren M, Rao GK, Scearce-Levie K, Schauer SP, Smith WB, Solanoy H, Teng E, Wildsmith KR, Bumbaca Yadav D, Ying Y, Fuji RN, Kerchner GA. Antibody semorinemab reduces tau pathology in a transgenic mouse model and engages tau in patients with Alzheimer's disease. Sci Transl Med 2021; 13:13/593/eabb2639. [PMID: 33980574 DOI: 10.1126/scitranslmed.abb2639] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 12/10/2020] [Indexed: 11/02/2022]
Abstract
Tau has become an attractive alternative target for passive immunotherapy efforts for Alzheimer's disease (AD). The anatomical distribution and extent of tau pathology correlate with disease course and severity better than other disease markers to date. We describe here the generation, preclinical characterization, and phase 1 clinical characterization of semorinemab, a humanized anti-tau monoclonal antibody with an immunoglobulin G4 (igG4) isotype backbone. Semorinemab binds all six human tau isoforms and protects neurons against tau oligomer neurotoxicity in cocultures of neurons and microglia. In addition, when administered intraperitoneally once weekly for 13 weeks, murine versions of semorinemab reduced the accumulation of tau pathology in a transgenic mouse model of tauopathy, independent of antibody effector function status. Semorinemab also showed clear evidence of target engagement in vivo, with increases in systemic tau concentrations observed in tau transgenic mice, nonhuman primates, and humans. Higher concentrations of systemic tau were observed after dosing in AD participants compared to healthy control participants. No concerning safety signals were observed in the phase 1 clinical trial at single doses up to 16,800 mg and multiple doses totaling 33,600 mg in a month.
Collapse
Affiliation(s)
- Gai Ayalon
- Department of Neuroscience, Genentech Inc., San Francisco, CA 94080, USA
| | - Seung-Hye Lee
- Department of Neuroscience, Genentech Inc., San Francisco, CA 94080, USA
| | - Oskar Adolfsson
- AC Immune SA, EPFL Innovation Park, Building B, CH-1015 Lausanne, Switzerland
| | | | - Jasvinder K Atwal
- Department of Neuroscience, Genentech Inc., San Francisco, CA 94080, USA
| | - Mira Blendstrup
- Department of Early Clinical Development, Genentech Inc., San Francisco, CA 94080, USA
| | - Helen Booler
- Department of Safety Assessment, Genentech Inc., San Francisco, CA 94080, USA
| | - Joseph Bravo
- Department of Safety Assessment, Genentech Inc., San Francisco, CA 94080, USA
| | - Robert Brendza
- Department of Neuroscience, Genentech Inc., San Francisco, CA 94080, USA
| | - Flavia Brunstein
- Department of Licensing and Early Development Safety, Genentech Inc., San Francisco, CA 94080, USA
| | - Ruby Chan
- Department of Protein Chemistry, Genentech Inc., San Francisco, CA 94080, USA
| | - Priya Chandra
- Department of Clinical Pharmacology, Genentech Inc., San Francisco, CA 94080, USA
| | - Jessica A Couch
- Project Team Leadership, Genentech Inc., San Francisco, CA 94080, USA
| | - Akash Datwani
- Department of Bioanalytical Sciences, Genentech Inc., San Francisco, CA 94080, USA
| | - Barthélemy Demeule
- Department of Late Stage Pharmaceutical Development, Genentech Inc., San Francisco, CA 94080, USA
| | - Danielle DiCara
- Department of Antibody Engineering, Genentech Inc., San Francisco, CA 94080, USA
| | - Rich Erickson
- Department of Bioanalytical Sciences, Genentech Inc., San Francisco, CA 94080, USA
| | - James A Ernst
- Department of Protein Chemistry, Genentech Inc., San Francisco, CA 94080, USA
| | - Oded Foreman
- Department of Pathology, Genentech Inc., San Francisco, CA 94080, USA
| | - Dongping He
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., San Francisco, CA 94080, USA
| | - Isidro Hötzel
- Department of Antibody Engineering, Genentech Inc., San Francisco, CA 94080, USA
| | - Michael Keeley
- Project Team Leadership, Genentech Inc., San Francisco, CA 94080, USA
| | - Michael C M Kwok
- Department of Protein Chemistry, Genentech Inc., San Francisco, CA 94080, USA
| | | | - Han Lin
- Department of Neuroscience, Genentech Inc., San Francisco, CA 94080, USA
| | - Yanmei Lu
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., San Francisco, CA 94080, USA
| | - Wilman Luk
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., San Francisco, CA 94080, USA
| | - Paul Manser
- Biostatistics, Genentech Inc., San Francisco, CA 94080, USA
| | - Andreas Muhs
- AC Immune SA, EPFL Innovation Park, Building B, CH-1015 Lausanne, Switzerland
| | - Hai Ngu
- Department of Pathology, Genentech Inc., San Francisco, CA 94080, USA
| | - Andrea Pfeifer
- AC Immune SA, EPFL Innovation Park, Building B, CH-1015 Lausanne, Switzerland
| | - Maria Pihlgren
- AC Immune SA, EPFL Innovation Park, Building B, CH-1015 Lausanne, Switzerland
| | - Gautham K Rao
- Department of Safety Assessment, Genentech Inc., San Francisco, CA 94080, USA
| | | | - Stephen P Schauer
- Department of Biomarker Development, Genentech Inc., San Francisco, CA 94080, USA
| | - William B Smith
- Alliance for Multispecialty Research, University of Tennessee Medical Center, Knoxville, TN 37920, USA
| | - Hilda Solanoy
- Department of Neuroscience, Genentech Inc., San Francisco, CA 94080, USA
| | - Edmond Teng
- Department of Early Clinical Development, Genentech Inc., San Francisco, CA 94080, USA
| | - Kristin R Wildsmith
- Department of Biomarker Development, Genentech Inc., San Francisco, CA 94080, USA
| | - Daniela Bumbaca Yadav
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., San Francisco, CA 94080, USA
| | - Yong Ying
- Department of Bioanalytical Sciences, Genentech Inc., San Francisco, CA 94080, USA
| | - Reina N Fuji
- Department of Safety Assessment, Genentech Inc., San Francisco, CA 94080, USA.
| | - Geoffrey A Kerchner
- Department of Early Clinical Development, Genentech Inc., San Francisco, CA 94080, USA
| |
Collapse
|
181
|
Ashton NJ, Leuzy A, Karikari TK, Mattsson-Carlgren N, Dodich A, Boccardi M, Corre J, Drzezga A, Nordberg A, Ossenkoppele R, Zetterberg H, Blennow K, Frisoni GB, Garibotto V, Hansson O. The validation status of blood biomarkers of amyloid and phospho-tau assessed with the 5-phase development framework for AD biomarkers. Eur J Nucl Med Mol Imaging 2021; 48:2140-2156. [PMID: 33677733 PMCID: PMC8175325 DOI: 10.1007/s00259-021-05253-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE The development of blood biomarkers that reflect Alzheimer's disease (AD) pathophysiology (phosphorylated tau and amyloid-β) has offered potential as scalable tests for dementia differential diagnosis and early detection. In 2019, the Geneva AD Biomarker Roadmap Initiative included blood biomarkers in the systematic validation of AD biomarkers. METHODS A panel of experts convened in November 2019 at a two-day workshop in Geneva. The level of maturity (fully achieved, partly achieved, preliminary evidence, not achieved, unsuccessful) of blood biomarkers was assessed based on the Biomarker Roadmap methodology and discussed fully during the workshop which also evaluated cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers. RESULTS Plasma p-tau has shown analytical validity (phase 2 primary aim 1) and first evidence of clinical validity (phase 3 primary aim 1), whereas the maturity level for Aβ remains to be partially achieved. Full and partial achievement has been assigned to p-tau and Aβ, respectively, in their associations to ante-mortem measures (phase 2 secondary aim 2). However, only preliminary evidence exists for the influence of covariates, assay comparison and cut-off criteria. CONCLUSIONS Despite the relative infancy of blood biomarkers, in comparison to CSF biomarkers, much has already been achieved for phases 1 through 3 - with p-tau having greater success in detecting AD and predicting disease progression. However, sufficient data about the effect of covariates on the biomarker measurement is lacking. No phase 4 (real-world performance) or phase 5 (assessment of impact/cost) aim has been tested, thus not achieved.
Collapse
Affiliation(s)
- N J Ashton
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden.
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - A Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - T K Karikari
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
| | - N Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - A Dodich
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Center for Neurocognitive Rehabilitation (CeRiN), CIMeC, University of Trento, Trento, Italy
| | - M Boccardi
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
| | - J Corre
- Centre National de la Recherche Scientifique, Montpellier, France
| | - A Drzezga
- Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - A Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital Stockholm, Stockholm, Sweden
| | - R Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - H Zetterberg
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - K Blennow
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - G B Frisoni
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - V Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Diagnostic Department, University Hospitals of Geneva, Geneva, Switzerland
| | - O Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- UK Dementia Research Institute at UCL, London, UK.
- Memory Clinic, Skåne University Hospital, SE-205 02, Malmö, Sweden.
| |
Collapse
|
182
|
Verde F, Otto M, Silani V. Neurofilament Light Chain as Biomarker for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Front Neurosci 2021; 15:679199. [PMID: 34234641 PMCID: PMC8255624 DOI: 10.3389/fnins.2021.679199] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/29/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two related currently incurable neurodegenerative diseases. ALS is characterized by degeneration of upper and lower motor neurons causing relentless paralysis of voluntary muscles, whereas in FTD, progressive atrophy of the frontal and temporal lobes of the brain results in deterioration of cognitive functions, language, personality, and behavior. In contrast to Alzheimer's disease (AD), ALS and FTD still lack a specific neurochemical biomarker reflecting neuropathology ex vivo. However, in the past 10 years, considerable progress has been made in the characterization of neurofilament light chain (NFL) as cerebrospinal fluid (CSF) and blood biomarker for both diseases. NFL is a structural component of the axonal cytoskeleton and is released into the CSF as a consequence of axonal damage or degeneration, thus behaving in general as a relatively non-specific marker of neuroaxonal pathology. However, in ALS, the elevation of its CSF levels exceeds that observed in most other neurological diseases, making it useful for the discrimination from mimic conditions and potentially worthy of consideration for introduction into diagnostic criteria. Moreover, NFL correlates with disease progression rate and is negatively associated with survival, thus providing prognostic information. In FTD patients, CSF NFL is elevated compared with healthy individuals and, to a lesser extent, patients with other forms of dementia, but the latter difference is not sufficient to enable a satisfying diagnostic performance at individual patient level. However, also in FTD, CSF NFL correlates with several measures of disease severity. Due to technological progress, NFL can now be quantified also in peripheral blood, where it is present at much lower concentrations compared with CSF, thus allowing less invasive sampling, scalability, and longitudinal measurements. The latter has promoted innovative studies demonstrating longitudinal kinetics of NFL in presymptomatic individuals harboring gene mutations causing ALS and FTD. Especially in ALS, NFL levels are generally stable over time, which, together with their correlation with progression rate, makes NFL an ideal pharmacodynamic biomarker for therapeutic trials. In this review, we illustrate the significance of NFL as biomarker for ALS and FTD and discuss unsolved issues and potential for future developments.
Collapse
Affiliation(s)
- Federico Verde
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Vincenzo Silani
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
183
|
Genome-wide CRISPR screen identifies protein pathways modulating tau protein levels in neurons. Commun Biol 2021; 4:736. [PMID: 34127790 PMCID: PMC8203616 DOI: 10.1038/s42003-021-02272-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 05/24/2021] [Indexed: 12/24/2022] Open
Abstract
Aggregates of hyperphosphorylated tau protein are a pathological hallmark of more than 20 distinct neurodegenerative diseases, including Alzheimer’s disease, progressive supranuclear palsy, and frontotemporal dementia. While the exact mechanism of tau aggregation is unknown, the accumulation of aggregates correlates with disease progression. Here we report a genome-wide CRISPR screen to identify modulators of endogenous tau protein for the first time. Primary screens performed in SH-SY5Y cells, identified positive and negative regulators of tau protein levels. Hit validation of the top 43 candidate genes was performed using Ngn2-induced human cortical excitatory neurons. Using this approach, genes and pathways involved in modulation of endogenous tau levels were identified, including chromatin modifying enzymes, neddylation and ubiquitin pathway members, and components of the mTOR pathway. TSC1, a critical component of the mTOR pathway, was further validated in vivo, demonstrating the relevance of this screening strategy. These findings may have implications for treating neurodegenerative diseases in the future. Using an unbiased genome-wide CRISPR screen approach, Sanchez et al. identified modulators of endogenous tau protein. This study suggests that chromatin modifiers, neddylation, ubiquitination, and the mTOR pathways regulate overall levels of tau protein in neurons, which could help in future identification of therapeutics for neurodegenerative diseases.
Collapse
|
184
|
Ji C, Sigurdsson EM. Current Status of Clinical Trials on Tau Immunotherapies. Drugs 2021; 81:1135-1152. [PMID: 34101156 DOI: 10.1007/s40265-021-01546-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/12/2022]
Abstract
Tau immunotherapies have advanced from proof-of-concept studies to over a dozen clinical trials for Alzheimer's disease (AD) and other tauopathies. Mechanistic studies in animal and culture models have provided valuable insight into how these therapies may work but multiple pathways are likely involved. Different groups have emphasized the importance of intracellular vs extracellular antibody-mediated clearance of the tau protein and there is no consensus on which pool of tau should ideally be targeted. Likewise, various normal and disease-selective epitopes are being targeted, and the antibody isotypes either favor phagocytosis of the tau-antibody complex or are neutral in that aspect. Most of the clinical trials are in early stages, thus their efficacy is not yet known, but all have been without any major adverse effects and some have reported target engagement. A few have been discontinued. One in phase I, presumably because of a poor pharmacokinetic profile, and three in phase II for a lack of efficacy although this trial stage is not well powered for efficacy measures. In these phase II studies, trials with two antibodies in patients with progressive supranuclear palsy or other primary tauopathies were halted but are continuing in patients with AD, and one antibody trial was stopped in early-stage AD but is continuing in moderate AD. These three antibodies have been reported to only work extracellularly and tau is not increased in the cerebrospinal fluid of primary tauopathies, which may explain the failures of two of them. In the discontinued AD trial, there are some concerns about how much of extracellular tau contains the N-terminal epitope that is being targeted. In addition, extracellular tau is only a small part of total tau, compared to intracellular tau. Targeting only the former may not be sufficient for functional benefits. Given these outcomes, decision makers within the pharmaceutical companies who green light these trials should attempt to target tau not only extracellularly but also intracellularly to increase their chances of success. Hopefully, some of the ongoing trials will provide some functional benefits to the large number of patients with tauopathies.
Collapse
Affiliation(s)
- Changyi Ji
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, Science Building, 11th floor, 435 East 30th Street, New York, NY, 10016, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, Science Building, 11th floor, 435 East 30th Street, New York, NY, 10016, USA. .,Department of Psychiatry, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
185
|
Pereira JB, Janelidze S, Stomrud E, Palmqvist S, van Westen D, Dage JL, Mattsson-Carlgren N, Hansson O. Plasma markers predict changes in amyloid, tau, atrophy and cognition in non-demented subjects. Brain 2021; 144:2826-2836. [PMID: 34077494 PMCID: PMC8557344 DOI: 10.1093/brain/awab163] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 11/13/2022] Open
Abstract
It is currently unclear whether plasma biomarkers can be used as independent prognostic tools to predict changes associated with early Alzheimer's disease (AD). In this study we sought to address this question by assessing whether plasma biomarkers can predict changes in amyloid load, tau accumulation, brain atrophy and cognition in non-demented individuals. To achieve this, plasma amyloid-β 42/40 (Aβ42/40), phosphorylated-tau181 (P-tau181), phosphorylated-tau217 (P-tau217) and neurofilament light (NfL) were determined in 159 non-demented individuals, 123 patients with AD dementia and 35 patients with a non-AD dementia from the Swedish BioFINDER-2 study, who underwent longitudinal amyloid (18 F-flutemetamol) and tau (18 F-RO948) positron emission tomography (PET), structural magnetic resonance imaging (T1-weighted) and cognitive testing. Our univariate linear mixed effect models showed there were several significant associations between the plasma biomarkers with imaging and cognitive measures. However, when all biomarkers were included in the same multivariate linear mixed effect models, we found that increased longitudinal amyloid-PET signals were independently predicted by low baseline plasma Aβ42/40 (p = 0.012), whereas increased tau-PET signals, brain atrophy and worse cognition were independently predicted by high plasma P-tau217 (p < 0.004). These biomarkers performed equally well or better than the corresponding biomarkers measured in the cerebrospinal fluid. In addition, they showed a similar performance to binary plasma biomarker values defined using the Youden index, which can be more easily implemented in the clinic. In addition, plasma Aβ42/40 and P-tau217 did not predict longitudinal changes in patients with a non-AD neurodegenerative disorder. In conclusion, our findings indicate that plasma Aβ42/40 and P-tau217 could be useful in clinical practice, research and drug development as prognostic markers of future AD pathology.
Collapse
Affiliation(s)
- Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, 141 83 Huddinge, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Memory Clinic, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Memory Clinic, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University, 221 85 Lund, Sweden.,Image and Function, Skåne University Hospital, Malmö 205 02, Sweden
| | | | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, 221 84 Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, SE-20502 Malmö, Sweden.,Memory Clinic, Skåne University Hospital, 214 28 Malmö, Sweden
| |
Collapse
|
186
|
Palmqvist S, Tideman P, Cullen N, Zetterberg H, Blennow K, Dage JL, Stomrud E, Janelidze S, Mattsson-Carlgren N, Hansson O. Prediction of future Alzheimer's disease dementia using plasma phospho-tau combined with other accessible measures. Nat Med 2021; 27:1034-1042. [PMID: 34031605 DOI: 10.1038/s41591-021-01348-z] [Citation(s) in RCA: 242] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/12/2021] [Indexed: 02/04/2023]
Abstract
A combination of plasma phospho-tau (P-tau) and other accessible biomarkers might provide accurate prediction about the risk of developing Alzheimer's disease (AD) dementia. We examined this in participants with subjective cognitive decline and mild cognitive impairment from the BioFINDER (n = 340) and Alzheimer's Disease Neuroimaging Initiative (ADNI) (n = 543) studies. Plasma P-tau, plasma Aβ42/Aβ40, plasma neurofilament light, APOE genotype, brief cognitive tests and an AD-specific magnetic resonance imaging measure were examined using progression to AD as outcome. Within 4 years, plasma P-tau217 predicted AD accurately (area under the curve (AUC) = 0.83) in BioFINDER. Combining plasma P-tau217, memory, executive function and APOE produced higher accuracy (AUC = 0.91, P < 0.001). In ADNI, this model had similar AUC (0.90) using plasma P-tau181 instead of P-tau217. The model was implemented online for prediction of the individual probability of progressing to AD. Within 2 and 6 years, similar models had AUCs of 0.90-0.91 in both cohorts. Using cerebrospinal fluid P-tau, Aβ42/Aβ40 and neurofilament light instead of plasma biomarkers did not improve the accuracy significantly. The clinical predictions by memory clinic physicians had significantly lower accuracy (4-year AUC = 0.71). In summary, plasma P-tau, in combination with brief cognitive tests and APOE genotyping, might greatly improve the diagnostic prediction of AD and facilitate recruitment for AD trials.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Nicholas Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at University College London, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | | | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
187
|
Bijttebier S, Theunis C, Jahouh F, Martins DR, Verhemeldonck M, Grauwen K, Dillen L, Mercken M. Development of immunoprecipitation - two-dimensional liquid chromatography - mass spectrometry methodology as biomarker read-out to quantify phosphorylated tau in cerebrospinal fluid from Alzheimer disease patients. J Chromatogr A 2021; 1651:462299. [PMID: 34107398 DOI: 10.1016/j.chroma.2021.462299] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
In Alzheimer's disease (AD) brain, one of the histopathological hallmarks is the neurofibrillary tangles consisting of aggregated and hyperphosphorylated tau. Currently many tau binding antibodies are under development to target the extracellular species responsible for the spreading of the disease in the brain. As such, an in-house developed antibody JNJ-63733657 with picomolar affinity towards tau phosphorylated at both T212 and T217 (further named p217+tau) was recently tested in phase I clinical trial NCT03375697. Following multiple dose administration in healthy subjects and subjects with AD, there were dose dependant reductions in free p217+tau fragments in cerebrospinal fluid (CSF) following antibody administration, as measured with a novel single molecule ELISA assay (Simoa PT3 x PT82 assay), demonstrating epitope engagement of the therapeutic antibody [Galpern, Haeverans, Janssens, Triana-Baltzer, Kolb, Li, Nandy, Mercken, Van Kolen, Sun, Van Nueten, 2020]. Total p217+tau levels also were reduced in CSF as measured with the Simoa PT3 x PT82 assay. In this study we developed an orthogonal immunoprecipitation - liquid chromatography - triple quadrupole mass spectrometry (IP-LC-TQMS) assay to verify the observed reductions in total p217+ tau levels. In this assay, an excess of JNJ-63733657 is added to the clinical CSF to ensure all p217+tau is bound by the antibody instead of having a pool of bound and unbound antigen and to immunoprecipitate all p217+tau, which is followed by on-bead digestion with trypsin to release surrogate peptides. Tryptic peptides with missed cleavages were monitored when phosphorylation occurred close to the cleavage site as this induced miscleavages. Compared with acidified mobile phases typically used for peptide analysis, reversed phase LC with mobile phase at basic pH resulted in sharper peaks and improved selectivity and sensitivity for the target peptides. With this setup a diphospho-tau tryptic peptide SRTPSLPTPPTREPK*2 could be measured with pT217 accounting for at least one of the phospho-sites. This is the first time that the presence of a diphopsho-tau peptide is reported to be present in human CSF. A two-dimensional LC-TQMS method was developed to remove matrix interferences. Selective trapping of diphospho-peptides via a metal oxide chromatography mechanism was achieved in a first dimension with a conventional reversed phase stationary phase and acidified mobile phase. Subsequent elution at basic pH enabled detection of low picomolar p217+tau levels in human CSF (lower limit of quantification: 2 pM), resulting in an approximate 5-fold increase in sensitivity. This enabled the quantification of total p217+tau in CSF leading to the confirmation that in addition to reductions in free p217+tau levels total p217+tau levels were also reduced following administration of the tau mAb JNJ-63733657, correlating with the previous measurement with the PT3 x PT82 Simoa assay. An orthogonal sample clean-up using offline TiO2/ZrO2 combined with 1DLC-TQMS was developed to confirm the presence of mono-ptau (pT217) tryptic peptides in CSF.
Collapse
Affiliation(s)
| | - Clara Theunis
- R&D Neurosciences, Janssen Pharmaceutica, Turnhoutseweg 30, Beerse, Belgium.
| | - Farid Jahouh
- DMPK, Janssen Pharmaceutica, Turnhoutseweg 30, Beerse, Belgium.
| | | | | | - Karolien Grauwen
- R&D Neurosciences, Janssen Pharmaceutica, Turnhoutseweg 30, Beerse, Belgium.
| | - Lieve Dillen
- DMPK, Janssen Pharmaceutica, Turnhoutseweg 30, Beerse, Belgium.
| | - Marc Mercken
- R&D Neurosciences, Janssen Pharmaceutica, Turnhoutseweg 30, Beerse, Belgium.
| |
Collapse
|
188
|
Triana‐Baltzer G, Moughadam S, Slemmon R, Van Kolen K, Theunis C, Mercken M, Kolb HC. Development and validation of a high-sensitivity assay for measuring p217+tau in plasma. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12204. [PMID: 34095436 PMCID: PMC8158165 DOI: 10.1002/dad2.12204] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Diagnosis of Alzheimer's disease (AD) based on amyloid beta (A), pathologic tau (T), and neurodegeneration (N) biomarkers in peripheral fluids promises to accelerate clinical trials and intercept disease earlier. METHODS Qualification of a Simoa plasma p217+tau assay was performed, followed by clinical utility evaluation in a cohort of 227 subjects with broad A and T spectrum. RESULTS The p217+tau plasma assay was accurate, precise, dilution linear, and highly sensitive. All measured samples were within linear range of the assay, presented higher concentration in AD versus healthy controls (P < .0001), and plasma and cerebrospinal fluid levels correlated (r2 = 0.35). The plasma p217+tau results were predictive of central T and A status (area under the curve = 0.90 and 0.90, respectively) with low false +/- rates. DISCUSSION The assay described here exhibits good technical performance and shows potential as a highly accurate peripheral biomarker for A or T status in AD and cognitively normal subjects.
Collapse
Affiliation(s)
| | - Setareh Moughadam
- Neuroscience BiomarkersJanssen Research & DevelopmentLa JollaCaliforniaUSA
| | - Randy Slemmon
- Neuroscience BiomarkersJanssen Research & DevelopmentLa JollaCaliforniaUSA
| | | | - Clara Theunis
- Neuroscience DepartmentJanssen Research & DevelopmentBeerseBelgium
| | - Marc Mercken
- Neuroscience DepartmentJanssen Research & DevelopmentBeerseBelgium
| | - Hartmuth C. Kolb
- Neuroscience BiomarkersJanssen Research & DevelopmentLa JollaCaliforniaUSA
| |
Collapse
|
189
|
Guo Y, Huang YY, Shen XN, Chen SD, Hu H, Wang ZT, Tan L, Yu JT. Characterization of Alzheimer's tau biomarker discordance using plasma, CSF, and PET. Alzheimers Res Ther 2021; 13:93. [PMID: 33947453 PMCID: PMC8094494 DOI: 10.1186/s13195-021-00834-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/21/2021] [Indexed: 12/03/2022]
Abstract
BACKGROUND We aimed to investigate the tau biomarker discrepancies of Alzheimer's disease (AD) using plasma tau phosphorylated at threonine 181 (p-tau181), cerebrospinal fluid (CSF) p-tau181, and AV1451 positron emission tomography (PET). METHODS In the Alzheimer's Disease Neuroimaging Initiative, 724 non-demented participants were categorized into plasma/CSF and plasma/PET groups. Demographic and clinical variables, amyloid-β (Aβ) burden, flortaucipir-PET binding in Braak regions of interest (ROIs), longitudinal changes in clinical outcomes, and conversion risk were compared. RESULTS Across different tau biomarker groups, the proportion of participants with a discordant profile varied (plasma+/CSF- 15.6%, plasma-/CSF+ 15.3%, plasma+/PET- 22.4%, and plasma-/PET+ 6.1%). Within the plasma/CSF categories, we found an increase from concordant-negative to discordant to concordant-positive in the frequency of Aβ pathology or cognitive impairment, rates of cognitive decline, and risk of cognitive conversion. However, the two discordant categories (plasma+/CSF- and plasma-/CSF+) showed comparable performances, resulting in similarly reduced cognitive capacities. Regarding plasma/PET categories, as expected, PET-positive individuals had increased Aβ burden, elevated flortaucipir retention in Braak ROIs, and accelerated cognitive deterioration than concordant-negative persons. Noteworthy, discordant participants with normal PET exhibited reduced flortaucipir uptake in Braak stage ROIs and slower rates of cognitive decline, relative to those PET-positive. Therefore, individuals with PET abnormality appeared to have advanced tau pathological changes and poorer cognitive function, regardless of the plasma status. Furthermore, these results were found only in individuals with Aβ pathology. CONCLUSIONS Our results indicate that plasma and CSF p-tau181 abnormalities associated with amyloidosis occur simultaneously in the progression of AD pathogenesis and related cognitive decline, before tau-PET turns positive.
Collapse
Affiliation(s)
- Yu Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu-Yuan Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
190
|
Ashton NJ, Pascoal TA, Karikari TK, Benedet AL, Lantero-Rodriguez J, Brinkmalm G, Snellman A, Schöll M, Troakes C, Hye A, Gauthier S, Vanmechelen E, Zetterberg H, Rosa-Neto P, Blennow K. Plasma p-tau231: a new biomarker for incipient Alzheimer's disease pathology. Acta Neuropathol 2021; 141:709-724. [PMID: 33585983 PMCID: PMC8043944 DOI: 10.1007/s00401-021-02275-6] [Citation(s) in RCA: 299] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/12/2021] [Accepted: 01/22/2021] [Indexed: 01/31/2023]
Abstract
The quantification of phosphorylated tau in biofluids, either cerebrospinal fluid (CSF) or plasma, has shown great promise in detecting Alzheimer's disease (AD) pathophysiology. Tau phosphorylated at threonine 231 (p-tau231) is one such biomarker in CSF but its usefulness as a blood biomarker is currently unknown. Here, we developed an ultrasensitive Single molecule array (Simoa) for the quantification of plasma p-tau231 which was validated in four independent cohorts (n = 588) in different settings, including the full AD continuum and non-AD neurodegenerative disorders. Plasma p-tau231 was able to identify patients with AD and differentiate them from amyloid-β negative cognitively unimpaired (CU) older adults with high accuracy (AUC = 0.92-0.94). Plasma p-tau231 also distinguished AD patients from patients with non-AD neurodegenerative disorders (AUC = 0.93), as well as from amyloid-β negative MCI patients (AUC = 0.89). In a neuropathology cohort, plasma p-tau231 in samples taken on avergae 4.2 years prior to post-mortem very accurately identified AD neuropathology in comparison to non-AD neurodegenerative disorders (AUC = 0.99), this is despite all patients being given an AD dementia diagnosis during life. Plasma p-tau231 was highly correlated with CSF p-tau231, tau pathology as assessed by [18F]MK-6240 positron emission tomography (PET), and brain amyloidosis by [18F]AZD469 PET. Remarkably, the inflection point of plasma p-tau231, increasing as a function of continuous [18F]AZD469 amyloid-β PET standardized uptake value ratio, was shown to be earlier than standard thresholds of amyloid-β PET positivity and the increase of plasma p-tau181. Furthermore, plasma p-tau231 was significantly increased in amyloid-β PET quartiles 2-4, whereas CSF p-tau217 and plasma p-tau181 increased only at quartiles 3-4 and 4, respectively. Finally, plasma p-tau231 differentiated individuals across the entire Braak stage spectrum, including Braak staging from Braak 0 through Braak I-II, which was not observed for plasma p-tau181. To conclude, this novel plasma p-tau231 assay identifies the clinical stages of AD and neuropathology equally well as plasma p-tau181, but increases earlier, already with subtle amyloid-β deposition, prior to the threshold for amyloid-β PET positivity has been attained, and also in response to early brain tau deposition. Thus, plasma p-tau231 is a promising novel biomarker of emerging AD pathology with the potential to facilitate clinical trials to identify vulnerable populations below PET threshold of amyloid-β positivity or apparent entorhinal tau deposition.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK.
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK.
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Department of Psychiatry and Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Abdul Hye
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Serge Gauthier
- Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, Montreal, McGill University, Montreal, QC, Canada
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Pedro Rosa-Neto
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| |
Collapse
|
191
|
Salvadó G, Milà‐Alomà M, Shekari M, Minguillon C, Fauria K, Niñerola‐Baizán A, Perissinotti A, Kollmorgen G, Buckley C, Farrar G, Zetterberg H, Blennow K, Suárez‐Calvet M, Molinuevo JL, Gispert JD. Cerebral amyloid-β load is associated with neurodegeneration and gliosis: Mediation by p-tau and interactions with risk factors early in the Alzheimer's continuum. Alzheimers Dement 2021; 17:788-800. [PMID: 33663013 PMCID: PMC8252618 DOI: 10.1002/alz.12245] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/06/2020] [Accepted: 10/24/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The association between cerebral amyloid-β accumulation and downstream CSF biomarkers is not fully understood, particularly in asymptomatic stages. METHODS In 318 cognitively unimpaired participants, we assessed the association between amyloid-β PET (Centiloid), and cerebrospinal fluid (CSF) biomarkers of several pathophysiological pathways. Interactions by Alzheimer's disease risk factors (age, sex and APOE-ε4), and the mediation effect of tau and neurodegeneration were also investigated. RESULTS Centiloids were positively associated with CSF biomarkers of tau pathology (p-tau), neurodegeneration (t-tau, NfL), synaptic dysfunction (neurogranin) and neuroinflammation (YKL-40, GFAP, sTREM2), presenting interactions with age (p-tau, t-tau, neurogranin) and sex (sTREM2, NfL). Most of these associations were mediated by p-tau, except for NfL. The interaction between sex and amyloid-β on sTREM2 and NfL was also tau-independent. DISCUSSION Early amyloid-β accumulation has a tau-independent effect on neurodegeneration and a tau-dependent effect on neuroinflammation. Besides, sex has a modifier effect on these associations independent of tau.
Collapse
Affiliation(s)
- Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | - Marta Milà‐Alomà
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Aida Niñerola‐Baizán
- Nuclear Medicine DepartmentHospital Clínic BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, (CIBER‐BBN)BarcelonaSpain
| | - Andrés Perissinotti
- Nuclear Medicine DepartmentHospital Clínic BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, (CIBER‐BBN)BarcelonaSpain
| | | | | | | | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, (CIBER‐BBN)BarcelonaSpain
| | | |
Collapse
|
192
|
Budelier MM, Bateman RJ. Biomarkers of Alzheimer Disease. J Appl Lab Med 2021; 5:194-208. [PMID: 31843944 DOI: 10.1373/jalm.2019.030080] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/31/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Alzheimer disease (AD) was once a clinical diagnosis confirmed by postmortem autopsy. Today, with the development of AD biomarkers, laboratory assays to detect AD pathology are able to complement clinical diagnosis in symptomatic individuals with uncertain diagnosis. A variety of commercially available assays are performed as laboratory-developed tests, and many more are in development for both clinical and research purposes. CONTENT The role of laboratory medicine in diagnosing and managing AD is expanding; thus, it is important for laboratory professionals and ordering physicians to understand the strengths and limitations of both existing and emerging AD biomarker assays. In this review, we will provide an overview of the diagnosis of AD, discuss existing laboratory assays for AD and their recommended use, and examine the clinical performance of emerging AD biomarkers. SUMMARY The field of AD biomarker discovery and assay development is rapidly evolving, with recent studies promising to improve both the diagnosis of symptomatic individuals and enrollment and monitoring of asymptomatic individuals in research studies. However, care must be taken to ensure proper use and interpretation of these assays. For clinical purposes, these assays are meant to aid in diagnosis but are not themselves diagnostic. For individuals without symptoms, AD biomarker tests are still only appropriate for research purposes. Additionally, there are analytical challenges that require careful attention, especially for longitudinal use of AD tests.
Collapse
Affiliation(s)
- Melissa M Budelier
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
193
|
Wegmann S, Biernat J, Mandelkow E. A current view on Tau protein phosphorylation in Alzheimer's disease. Curr Opin Neurobiol 2021; 69:131-138. [PMID: 33892381 DOI: 10.1016/j.conb.2021.03.003] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
The functions of the neuronal microtubule-associated protein Tau in the central nervous system are regulated by manifold posttranslational modifications at more than 50 sites. Tau in healthy neurons carries multiple phosphate groups, mostly in its microtubule assembly domain. Elevated phosphorylation and aggregation of Tau are widely considered pathological hallmarks in Alzheimer's disease (AD) and other tauopathies, triggering the quest for Tau posttranslational modifications in the disease context. However, the phosphorylation patterns of physiological and pathological Tau are surprisingly similar and heterogenous, making it difficult to identify specific modifications as therapeutic targets and biomarkers for AD. We present a concise summary of - and view on - important previous and recent advances in Tau phosphorylation analysis in the context of AD.
Collapse
Affiliation(s)
- Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.
| | - Jacek Biernat
- German Center for Neurodegenerative Diseases (DZNE) & CAESAR Research Center, Bonn, Germany
| | - Eckhard Mandelkow
- German Center for Neurodegenerative Diseases (DZNE) & CAESAR Research Center, Bonn, Germany
| |
Collapse
|
194
|
Pereira JB, Janelidze S, Ossenkoppele R, Kvartsberg H, Brinkmalm A, Mattsson-Carlgren N, Stomrud E, Smith R, Zetterberg H, Blennow K, Hansson O. Untangling the association of amyloid-β and tau with synaptic and axonal loss in Alzheimer's disease. Brain 2021; 144:310-324. [PMID: 33279949 PMCID: PMC8210638 DOI: 10.1093/brain/awaa395] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/04/2020] [Accepted: 09/21/2020] [Indexed: 01/10/2023] Open
Abstract
It is currently unclear how amyloid-β and tau deposition are linked to changes in
synaptic function and axonal structure over the course of Alzheimer’s disease. Here, we
assessed these relationships by measuring presynaptic (synaptosomal-associated protein 25,
SNAP25; growth-associated protein 43, GAP43), postsynaptic (neurogranin, NRGN) and axonal
(neurofilament light chain) markers in the CSF of individuals with varying levels of
amyloid-β and tau pathology based on 18F-flutemetamol PET and
18F-flortaucipir PET. In addition, we explored the relationships between
synaptic and axonal markers with cognition as well as functional and anatomical brain
connectivity markers derived from resting-state functional MRI and diffusion tensor
imaging. We found that the presynaptic and postsynaptic markers SNAP25, GAP43 and NRGN are
elevated in early Alzheimer’s disease i.e. in amyloid-β-positive individuals without
evidence of tau pathology. These markers were associated with greater amyloid-β pathology,
worse memory and functional changes in the default mode network. In contrast,
neurofilament light chain was abnormal in later disease stages, i.e. in individuals with
both amyloid-β and tau pathology, and correlated with more tau and worse global cognition.
Altogether, these findings support the hypothesis that amyloid-β and tau might have
differential downstream effects on synaptic and axonal function in a stage-dependent
manner, with amyloid-related synaptic changes occurring first, followed by tau-related
axonal degeneration.
Collapse
Affiliation(s)
- Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Hlin Kvartsberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
195
|
Hays CC, Zlatar ZZ, Meloy MJ, Osuna J, Liu TT, Galasko DR, Wierenga CE. Anterior Cingulate Structure and Perfusion is Associated with Cerebrospinal Fluid Tau among Cognitively Normal Older Adult APOEɛ4 Carriers. J Alzheimers Dis 2021; 73:87-101. [PMID: 31743999 DOI: 10.3233/jad-190504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Evidence suggests the ɛ4 allele of the apolipoprotein E (APOE) gene may accelerate an age-related process of cortical thickening and cerebral blood flow (CBF) reduction in the anterior cingulate cortex (ACC). Although the neural basis of this association remains unclear, evidence suggests it might reflect early neurodegenerative processes. However, to date, associations between cerebrospinal fluid (CSF) biomarkers of neurodegeneration, such as CSF tau, and APOE-related alterations in ACC cortical thickness (CTH) and CBF have yet to be explored. The current study explored the interaction of CSF tau and APOE genotype (ɛ4+, ɛ4-) on FreeSurfer-derived CTH and arterial spin labeling MRI-measured resting CBF in the ACC (caudal ACC [cACC] and rostral ACC [rACC]) among a sample of 45 cognitively normal older adults. Secondary analyses also examined associations between APOE, CTH/CBF, and cognitive performance. In the cACC, higher CSF tau was associated with higher CTH and lower CBF in ɛ4+, whereas these relationships were not evident in ɛ4-. In the rACC, higher CSF tau was associated with higher CTH for both ɛ4+ and ɛ4-, and with lower CBF only in ɛ4+. Significant interactions of CSF tau and APOE on CTH/CBF were not observed in two posterior reference regions implicated in Alzheimer's disease. Secondary analyses revealed a negative relationship between cACC CTH and executive functioning in ɛ4+ and a positive relationship in ɛ4-. Findings suggest the presence of an ɛ4-related pattern of increased CTH and reduced CBF in the ACC that is associated with biomarkers of neurodegeneration and subtle decrements in cognition.
Collapse
Affiliation(s)
- Chelsea C Hays
- VA San Diego Healthcare System, San Diego, CA, USA.,SDSU/UC San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Zvinka Z Zlatar
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA.,SDSU/UC San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - M J Meloy
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Jessica Osuna
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | - Thomas T Liu
- Department of Radiology, UC San Diego, La Jolla, CA, USA
| | - Douglas R Galasko
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Neurosciences, UC San Diego, La Jolla, CA, USA
| | - Christina E Wierenga
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, UC San Diego, La Jolla, CA, USA.,SDSU/UC San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| |
Collapse
|
196
|
Horie K, Barthélemy NR, Sato C, Bateman RJ. CSF tau microtubule binding region identifies tau tangle and clinical stages of Alzheimer's disease. Brain 2021; 144:515-527. [PMID: 33283854 DOI: 10.1093/brain/awaa373] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/08/2020] [Accepted: 08/19/2020] [Indexed: 12/14/2022] Open
Abstract
Tau is a microtubule associated protein in the brain that aggregates in Alzheimer's disease to form pathological tangles and neurites. Insoluble tau aggregates composed of the microtubule binding region (MTBR) of tau are highly associated with the cognitive and clinical symptoms of Alzheimer's disease. In contrast, levels of soluble forms of tau, such as CSF total tau and phosphorylated tau-181 and tau-217, increase prior to tau aggregation in Alzheimer's disease, but these biomarkers do not measure the MTBR of tau. Thus, how CSF MTBR-tau is altered in Alzheimer's disease remains unclear. In this study, we used sequential immunoprecipitation and chemical extraction methods followed by mass spectrometry to analyse MTBR-tau species in Alzheimer's disease and control CSF. We quantified MTBR-tau-specific regions in the CSF and identified that species containing the region beginning at residue 243 were the most highly correlated with tau PET and cognitive measures. This finding suggests that CSF level of tau species containing the upstream region of MTBR may reflect changes in tau pathology that occur in Alzheimer's disease and could serve as biomarkers to stage Alzheimer's disease and track the development of tau-directed therapeutics.
Collapse
Affiliation(s)
- Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
197
|
Tau Is Truncated in Five Regions of the Normal Adult Human Brain. Int J Mol Sci 2021; 22:ijms22073521. [PMID: 33805376 PMCID: PMC8036332 DOI: 10.3390/ijms22073521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 01/01/2023] Open
Abstract
The truncation of Tau is thought to be important in promoting aggregation, with this feature characterising the pathology of dementias such as Alzheimer disease. Antibodies to the C-terminal and N-terminal regions of Tau were employed to examine Tau cleavage in five human brain regions: the entorhinal cortex, prefrontal cortex, motor cortex, hippocampus, and cerebellum. These were obtained from normal subjects ranging in age from 18 to 104 years. Tau fragments of approximately 40 kDa and 45 kDa with an intact N-terminus retained were found in soluble and insoluble brain fractions. In addition, smaller C-terminal Tau fragments ranging in mass from 17 kDa to 25 kDa were also detected. These findings are consistent with significant Tau cleavage taking place in brain regions from 18 years onwards. It appears that site-specific cleavage of Tau is widespread in the normal human brain, and that large Tau fragments that contain the N-terminus, as well as shorter C-terminal Tau fragments, are present in brain cells across the age range.
Collapse
|
198
|
Carlomagno Y, Manne S, DeTure M, Prudencio M, Zhang YJ, Hanna Al-Shaikh R, Dunmore JA, Daughrity LM, Song Y, Castanedes-Casey M, Lewis-Tuffin LJ, Nicholson KA, Wszolek ZK, Dickson DW, Fitzpatrick AWP, Petrucelli L, Cook CN. The AD tau core spontaneously self-assembles and recruits full-length tau to filaments. Cell Rep 2021; 34:108843. [PMID: 33730588 PMCID: PMC8094113 DOI: 10.1016/j.celrep.2021.108843] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/18/2020] [Accepted: 02/17/2021] [Indexed: 02/05/2023] Open
Abstract
Tau accumulation is a major pathological hallmark of Alzheimer's disease (AD) and other tauopathies, but the mechanism(s) of tau aggregation remains unclear. Taking advantage of the identification of tau filament cores by cryoelectron microscopy, we demonstrate that the AD tau core possesses the intrinsic ability to spontaneously aggregate in the absence of an inducer, with antibodies generated against AD tau core filaments detecting AD tau pathology. The AD tau core also drives aggregation of full-length wild-type tau, increases seeding potential, and templates abnormal forms of tau present in brain homogenates and antemortem cerebrospinal fluid (CSF) from patients with AD in an ultrasensitive real-time quaking-induced conversion (QuIC) assay. Finally, we show that the filament cores in corticobasal degeneration (CBD) and Pick's disease (PiD) similarly assemble into filaments under physiological conditions. These results document an approach to modeling tau aggregation and have significant implications for in vivo investigation of tau transmission and biomarker development.
Collapse
Affiliation(s)
- Yari Carlomagno
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sireesha Manne
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Mercedes Prudencio
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | | | | | | | - Yuping Song
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Katharine A Nicholson
- Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital (MGH), Boston, MA, USA
| | | | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Anthony W P Fitzpatrick
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
| | - Casey N Cook
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
| |
Collapse
|
199
|
Wang W, Zhou Q, Jiang T, Li S, Ye J, Zheng J, Wang X, Liu Y, Deng M, Ke D, Wang Q, Wang Y, Wang JZ. A novel small-molecule PROTAC selectively promotes tau clearance to improve cognitive functions in Alzheimer-like models. Theranostics 2021; 11:5279-5295. [PMID: 33859747 PMCID: PMC8039949 DOI: 10.7150/thno.55680] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/18/2021] [Indexed: 12/21/2022] Open
Abstract
Intracellular accumulation of tau is a hallmark pathology in Alzheimer disease (AD) and the related tauopathies, thus targeting tau could be promising for drug development. Proteolysis Targeting Chimera (PROTAC) is a novel drug discovery strategy for selective protein degradation from within cells. Methods: A novel small-molecule PROTAC, named as C004019 with a molecular mass of 1,035.29 dalton, was designed to simultaneously recruite tau and E3-ligase (Vhl) and thus to selectively enhance ubiquitination and proteolysis of tau proteins. Western blotting, immunofluoresence and immunohistochemical staining were employed to verify the effects of C004019 in cell models (HEK293 and SH-SY5Y) and mouse models (hTau-transgenic and 3xTg-AD), respectively. The cognitive capacity of the mice was assessed by a suite of behavior experiments. Electrophysiology and Golgi staining were used to evaluate the synaptic plasticity. Results: C004019 induced a robust tau clearance via promoting its ubiquitination-proteasome-dependent proteolysis in HEK293 cells with stable or transient overexpression of human tau (hTau), and in SH-SY5Y that constitutively overexpress hTau. Furthermore, intracerebral ventricular infusion of C004019 induced a robust tau clearance in vivo. Most importantly, both single-dose and multiple-doses (once per 6 days for a total 5 times) subcutaneous administration of C004019 remarkably decreased tau levels in the brains of wild-type, hTau-transgenic and 3xTg-AD mice with improvement of synaptic and cognitive functions. Conclusions: The PROTAC (C004019) created in the current study can selectively and efficiently promote tau clearance both in vitro and in vivo, which provides a promising drug candidate for AD and the related tauopathies.
Collapse
Affiliation(s)
- Weijin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiuzhi Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shihong Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinwang Ye
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jie Zheng
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanchao Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Minmin Deng
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yipeng Wang
- Neurosmart Therapeutics Co., Ltd., Room 5013, Unit 1, Buiilding 7, Basheng road 160, Shanghai 200131, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China
| |
Collapse
|
200
|
Barthélemy NR, Horie K, Sato C, Bateman RJ. Blood plasma phosphorylated-tau isoforms track CNS change in Alzheimer's disease. J Exp Med 2021; 217:151982. [PMID: 32725127 PMCID: PMC7596823 DOI: 10.1084/jem.20200861] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 01/31/2023] Open
Abstract
Highly sensitive and specific plasma biomarkers for Alzheimer’s disease (AD) have the potential to improve diagnostic accuracy in the clinic and facilitate research studies including enrollment in prevention and treatment trials. We recently reported CSF tau hyperphosphorylation, especially on T217, is an accurate predictor of β-amyloidosis at asymptomatic and symptomatic stages. In the current study, we determine by mass spectrometry the potential utility of plasma p-tau isoforms to detect AD pathology and investigate CSF and plasma tau isoforms’ profile relationships. Plasma tau was truncated as previously described in CSF. CSF and plasma measures of p-tau-217 and p-tau-181 were correlated. No correlation was found between CSF and plasma on total-tau levels and pS202 measures. We found p-tau-217 and p-tau-181 were highly specific for amyloid plaque pathology in the discovery cohort (n = 36, AUROC = 0.99 and 0.98 respectively). In the validation cohort (n = 92), p-tau-217 measures were still specific to amyloid status (AUROC = 0.92), and p-tau-181 measures were less specific (AUROC = 0.75).
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|