1
|
Diez I, Ortiz-Terán L, Ng TSC, Albers MW, Marshall G, Orwig W, Kim CM, Bueichekú E, Montal V, Olofsson J, Vannini P, El Fahkri G, Sperling R, Johnson K, Jacobs HIL, Sepulcre J. Tau propagation in the brain olfactory circuits is associated with smell perception changes in aging. Nat Commun 2024; 15:4809. [PMID: 38844444 PMCID: PMC11156945 DOI: 10.1038/s41467-024-48462-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
The direct access of olfactory afferents to memory-related cortical systems has inspired theories about the role of the olfactory pathways in the development of cortical neurodegeneration in Alzheimer's disease (AD). In this study, we used baseline olfactory identification measures with longitudinal flortaucipir and PiB PET, diffusion MRI of 89 cognitively normal older adults (73.82 ± 8.44 years; 56% females), and a transcriptomic data atlas to investigate the spatiotemporal spreading and genetic vulnerabilities of AD-related pathology aggregates in the olfactory system. We find that odor identification deficits are predominantly associated with tau accumulation in key areas of the olfactory pathway, with a particularly strong predictive power for longitudinal tau progression. We observe that tau spreads from the medial temporal lobe structures toward the olfactory system, not the reverse. Moreover, we observed a genetic background of odor perception-related genes that might confer vulnerability to tau accumulation along the olfactory system.
Collapse
Affiliation(s)
- Ibai Diez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Laura Ortiz-Terán
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- UMASS Memorial Medical Center, UMASS Chan Medical School, Worcester, MA, USA
| | - Thomas S C Ng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark W Albers
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Gad Marshall
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - William Orwig
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Harvard University, Department of Psychology, Cambridge, MA, USA
| | - Chan-Mi Kim
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elisenda Bueichekú
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor Montal
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Barcelona Supercomputing Center, Barcelona, Spain
| | - Jonas Olofsson
- Stockholm University, Department of Psychology, Stockholm, Sweden
| | - Patrizia Vannini
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Georges El Fahkri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa Sperling
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith Johnson
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jorge Sepulcre
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
2
|
Doering S, McCullough A, Gordon BA, Chen CD, McKay N, Hobbs D, Keefe S, Flores S, Scott J, Smith H, Jarman S, Jackson K, Hornbeck RC, Ances BM, Xiong C, Aschenbrenner AJ, Hassenstab J, Cruchaga C, Daniels A, Bateman RJ, Morris JC, Benzinger TLS. Deconstructing pathological tau by biological process in early stages of Alzheimer disease: a method for quantifying tau spatial spread in neuroimaging. EBioMedicine 2024; 103:105080. [PMID: 38552342 PMCID: PMC10995809 DOI: 10.1016/j.ebiom.2024.105080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Neuroimaging studies often quantify tau burden in standardized brain regions to assess Alzheimer disease (AD) progression. However, this method ignores another key biological process in which tau spreads to additional brain regions. We have developed a metric for calculating the extent tau pathology has spread throughout the brain and evaluate the relationship between this metric and tau burden across early stages of AD. METHODS 445 cross-sectional participants (aged ≥ 50) who had MRI, amyloid PET, tau PET, and clinical testing were separated into disease-stage groups based on amyloid positivity and cognitive status (older cognitively normal control, preclinical AD, and symptomatic AD). Tau burden and tau spatial spread were calculated for all participants. FINDINGS We found both tau metrics significantly elevated across increasing disease stages (p < 0.0001) and as a function of increasing amyloid burden for participants with preclinical (p < 0.0001, p = 0.0056) and symptomatic (p = 0.010, p = 0.0021) AD. An interaction was found between tau burden and tau spatial spread when predicting amyloid burden (p = 0.00013). Analyses of slope between tau metrics demonstrated more spread than burden in preclinical AD (β = 0.59), but then tau burden elevated relative to spread (β = 0.42) once participants had symptomatic AD, when the tau metrics became highly correlated (R = 0.83). INTERPRETATION Tau burden and tau spatial spread are both strong biomarkers for early AD but provide unique information, particularly at the preclinical stage. Tau spatial spread may demonstrate earlier changes than tau burden which could have broad impact in clinical trial design. FUNDING This research was supported by the Knight Alzheimer Disease Research Center (Knight ADRC, NIH grants P30AG066444, P01AG026276, P01AG003991), Dominantly Inherited Alzheimer Network (DIAN, NIH grants U01AG042791, U19AG03243808, R01AG052550-01A1, R01AG05255003), and the Barnes-Jewish Hospital Foundation Willman Scholar Fund.
Collapse
Affiliation(s)
- Stephanie Doering
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Nicole McKay
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Diana Hobbs
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Sarah Keefe
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jalen Scott
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Hunter Smith
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Stephen Jarman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Kelley Jackson
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Beau M Ances
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Chengjie Xiong
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | | - Jason Hassenstab
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Alisha Daniels
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Randall J Bateman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | |
Collapse
|
3
|
Wisch JK, McKay NS, Boerwinkle AH, Kennedy J, Flores S, Handen BL, Christian BT, Head E, Mapstone M, Rafii MS, O'Bryant SE, Price JC, Laymon CM, Krinsky-McHale SJ, Lai F, Rosas HD, Hartley SL, Zaman S, Lott IT, Tudorascu D, Zammit M, Brickman AM, Lee JH, Bird TD, Cohen A, Chrem P, Daniels A, Chhatwal JP, Cruchaga C, Ibanez L, Jucker M, Karch CM, Day GS, Lee JH, Levin J, Llibre-Guerra J, Li Y, Lopera F, Roh JH, Ringman JM, Supnet-Bell C, van Dyck CH, Xiong C, Wang G, Morris JC, McDade E, Bateman RJ, Benzinger TLS, Gordon BA, Ances BM. Comparison of tau spread in people with Down syndrome versus autosomal-dominant Alzheimer's disease: a cross-sectional study. Lancet Neurol 2024; 23:500-510. [PMID: 38631766 PMCID: PMC11209765 DOI: 10.1016/s1474-4422(24)00084-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/01/2024] [Accepted: 02/21/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND In people with genetic forms of Alzheimer's disease, such as in Down syndrome and autosomal-dominant Alzheimer's disease, pathological changes specific to Alzheimer's disease (ie, accumulation of amyloid and tau) occur in the brain at a young age, when comorbidities related to ageing are not present. Studies including these cohorts could, therefore, improve our understanding of the early pathogenesis of Alzheimer's disease and be useful when designing preventive interventions targeted at disease pathology or when planning clinical trials. We compared the magnitude, spatial extent, and temporal ordering of tau spread in people with Down syndrome and autosomal-dominant Alzheimer's disease. METHODS In this cross-sectional observational study, we included participants (aged ≥25 years) from two cohort studies. First, we collected data from the Dominantly Inherited Alzheimer's Network studies (DIAN-OBS and DIAN-TU), which include carriers of autosomal-dominant Alzheimer's disease genetic mutations and non-carrier familial controls recruited in Australia, Europe, and the USA between 2008 and 2022. Second, we collected data from the Alzheimer Biomarkers Consortium-Down Syndrome study, which includes people with Down syndrome and sibling controls recruited from the UK and USA between 2015 and 2021. Controls from the two studies were combined into a single group of familial controls. All participants had completed structural MRI and tau PET (18F-flortaucipir) imaging. We applied Gaussian mixture modelling to identify regions of high tau PET burden and regions with the earliest changes in tau binding for each cohort separately. We estimated regional tau PET burden as a function of cortical amyloid burden for both cohorts. Finally, we compared the temporal pattern of tau PET burden relative to that of amyloid. FINDINGS We included 137 people with Down syndrome (mean age 38·5 years [SD 8·2], 74 [54%] male, and 63 [46%] female), 49 individuals with autosomal-dominant Alzheimer's disease (mean age 43·9 years [11·2], 22 [45%] male, and 27 [55%] female), and 85 familial controls, pooled from across both studies (mean age 41·5 years [12·1], 28 [33%] male, and 57 [67%] female), who satisfied the PET quality-control procedure for tau-PET imaging processing. 134 (98%) people with Down syndrome, 44 (90%) with autosomal-dominant Alzheimer's disease, and 77 (91%) controls also completed an amyloid PET scan within 3 years of tau PET imaging. Spatially, tau PET burden was observed most frequently in subcortical and medial temporal regions in people with Down syndrome, and within the medial temporal lobe in people with autosomal-dominant Alzheimer's disease. Across the brain, people with Down syndrome had greater concentrations of tau for a given level of amyloid compared with people with autosomal-dominant Alzheimer's disease. Temporally, increases in tau were more strongly associated with increases in amyloid for people with Down syndrome compared with autosomal-dominant Alzheimer's disease. INTERPRETATION Although the general progression of amyloid followed by tau is similar for people Down syndrome and people with autosomal-dominant Alzheimer's disease, we found subtle differences in the spatial distribution, timing, and magnitude of the tau burden between these two cohorts. These differences might have important implications; differences in the temporal pattern of tau accumulation might influence the timing of drug administration in clinical trials, whereas differences in the spatial pattern and magnitude of tau burden might affect disease progression. FUNDING None.
Collapse
Affiliation(s)
- Julie K Wisch
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA.
| | - Nicole S McKay
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Anna H Boerwinkle
- McGovern Medical School, University of Texas in Houston, Houston, TX, USA
| | - James Kennedy
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Shaney Flores
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Benjamin L Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley T Christian
- Department of Medical Physics and Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth Head
- Department of Pathology, Gillespie Neuroscience Research Facility, University of California, Irvine, CA, USA
| | - Mark Mapstone
- Department of Neurology, University of California Irvine School of Medicine, Irvine, CA, USA
| | - Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Sid E O'Bryant
- Institute for Translational Research Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Julie C Price
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sharon J Krinsky-McHale
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, USA
| | - Florence Lai
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - H Diana Rosas
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sigan L Hartley
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Shahid Zaman
- Cambridge Intellectual and Developmental Disabilities Research Group, University of Cambridge, Cambridge, UK
| | - Ira T Lott
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA, USA
| | - Dana Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew Zammit
- Department of Medical Physics and Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam M Brickman
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joseph H Lee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Thomas D Bird
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Annie Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricio Chrem
- Centro de Memoria y Envejecimiento, Buenos Aires, Argentina
| | - Alisha Daniels
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Celeste M Karch
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asian Medical Center, Seoul, South Korea
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases, site Munich, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| | - Jorge Llibre-Guerra
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Jee Hoon Roh
- Departments of Physiology and Neurology, Korea University College of Medicine, Seoul, South Korea
| | - John M Ringman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | | | | | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Guoqiao Wang
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | | | - Brian A Gordon
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|
4
|
Weinstein G, Kojis DJ, Ghosh S, Beiser AS, Seshadri S. Association of Neurotrophic Factors at Midlife With In Vivo Measures of β-Amyloid and Tau Burden 15 Years Later in Dementia-Free Adults. Neurology 2024; 102:e209198. [PMID: 38471064 PMCID: PMC11033983 DOI: 10.1212/wnl.0000000000209198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/13/2023] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Neurotrophic factors (NTFs) play an important role in Alzheimer disease (AD) pathophysiology. Brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) are important NTFs. However, a direct link of BDNF and VEGF circulating levels with in vivo measures of amyloid-β (Aβ) and tau burden remains to be elucidated. We explored the relationship of BDNF and VEGF serum levels with future brain Aβ and tau pathology in a cohort of cognitively healthy, predominantly middle-aged adults and tested for possible effect modifications by sex and menopausal status. METHODS This cross-sectional analysis was conducted using data from the Framingham Heart Study (FHS), a community-based cohort study. The study sample included cognitively healthy participants from the FHS Offspring and Third-generation cohorts. BDNF and VEGF were measured in the third-generation cohort during examination cycles 2 (2005-2008) and 1 (2002-2005), respectively, and in the offspring cohort during examination cycle 7 (1998-2001). Participants underwent 11C-Pittsburgh compound B amyloid and 18F-Flortaucipir tau-PET imaging (2015-2021). Linear regression models were used to assess the relationship of serum BDNF and VEGF levels with regional tau and global Aβ, adjusting for potential confounders. Interactions with sex and menopausal status were additionally tested. RESULTS The sample included 414 individuals (mean age = 41 ± 9 years; 51% female). Continuous measures of BDNF and VEGF were associated with tau signal in the rhinal region after adjustment for potential confounders (β = -0.15 ± 0.06, p = 0.018 and β = -0.19 ± 0.09, p = 0.043, respectively). High BDNF (≥32,450 pg/mL) and VEGF (≥488 pg/mL) levels were significantly related to lower rhinal tau (β = -0.27 ± 0.11, p = 0.016 and β = -0.40 ± 0.14, p = 0.004, respectively) and inferior temporal tau (β = -0.24 ± 0.11, p = 0.028 and β = -0.26 ± 0.13, p = 0.049, respectively). The BDNF-rhinal tau association was observed only among male individuals. Overall, BDNF and VEGF were not associated with global amyloid; however, high VEGF levels were associated with lower amyloid burden in postmenopausal women (β = -1.96 ± 0.70, p = 0.013, per 1 pg/mL). DISCUSSION This study demonstrates a robust association between BDNF and VEGF serum levels with in vivo measures of tau almost 2 decades later. These findings add to mounting evidence from preclinical studies suggesting a role of NTFs as valuable blood biomarkers for AD risk prediction.
Collapse
Affiliation(s)
- Galit Weinstein
- From the School of Public Health (G.W.), University of Haifa, Israel; Department of Biostatistics (D.J.K., A.S.B.), Boston University School of Public Health, Boston; The Framingham Study (D.J.K., S.G., A.S.B., S.S.); Department of Neurology (S.G., A.S.B., S.S.), Boston University Chobanian & Avedisian School of Medicine, MA; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio
| | - Daniel J Kojis
- From the School of Public Health (G.W.), University of Haifa, Israel; Department of Biostatistics (D.J.K., A.S.B.), Boston University School of Public Health, Boston; The Framingham Study (D.J.K., S.G., A.S.B., S.S.); Department of Neurology (S.G., A.S.B., S.S.), Boston University Chobanian & Avedisian School of Medicine, MA; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio
| | - Saptaparni Ghosh
- From the School of Public Health (G.W.), University of Haifa, Israel; Department of Biostatistics (D.J.K., A.S.B.), Boston University School of Public Health, Boston; The Framingham Study (D.J.K., S.G., A.S.B., S.S.); Department of Neurology (S.G., A.S.B., S.S.), Boston University Chobanian & Avedisian School of Medicine, MA; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio
| | - Alexa S Beiser
- From the School of Public Health (G.W.), University of Haifa, Israel; Department of Biostatistics (D.J.K., A.S.B.), Boston University School of Public Health, Boston; The Framingham Study (D.J.K., S.G., A.S.B., S.S.); Department of Neurology (S.G., A.S.B., S.S.), Boston University Chobanian & Avedisian School of Medicine, MA; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio
| | - Sudha Seshadri
- From the School of Public Health (G.W.), University of Haifa, Israel; Department of Biostatistics (D.J.K., A.S.B.), Boston University School of Public Health, Boston; The Framingham Study (D.J.K., S.G., A.S.B., S.S.); Department of Neurology (S.G., A.S.B., S.S.), Boston University Chobanian & Avedisian School of Medicine, MA; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (S.S.), University of Texas Health Sciences Center, San Antonio
| |
Collapse
|
5
|
Meeker KL, Luckett PH, Barthélemy NR, Hobbs DA, Chen C, Bollinger J, Ovod V, Flores S, Keefe S, Henson RL, Herries EM, McDade E, Hassenstab JJ, Xiong C, Cruchaga C, Benzinger TLS, Holtzman DM, Schindler SE, Bateman RJ, Morris JC, Gordon BA, Ances BM. Comparison of cerebrospinal fluid, plasma and neuroimaging biomarker utility in Alzheimer's disease. Brain Commun 2024; 6:fcae081. [PMID: 38505230 PMCID: PMC10950051 DOI: 10.1093/braincomms/fcae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/01/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
Alzheimer's disease biomarkers are crucial to understanding disease pathophysiology, aiding accurate diagnosis and identifying target treatments. Although the number of biomarkers continues to grow, the relative utility and uniqueness of each is poorly understood as prior work has typically calculated serial pairwise relationships on only a handful of markers at a time. The present study assessed the cross-sectional relationships among 27 Alzheimer's disease biomarkers simultaneously and determined their ability to predict meaningful clinical outcomes using machine learning. Data were obtained from 527 community-dwelling volunteers enrolled in studies at the Charles F. and Joanne Knight Alzheimer Disease Research Center at Washington University in St Louis. We used hierarchical clustering to group 27 imaging, CSF and plasma measures of amyloid beta, tau [phosphorylated tau (p-tau), total tau t-tau)], neuronal injury and inflammation drawn from MRI, PET, mass-spectrometry assays and immunoassays. Neuropsychological and genetic measures were also included. Random forest-based feature selection identified the strongest predictors of amyloid PET positivity across the entire cohort. Models also predicted cognitive impairment across the entire cohort and in amyloid PET-positive individuals. Four clusters emerged reflecting: core Alzheimer's disease pathology (amyloid and tau), neurodegeneration, AT8 antibody-associated phosphorylated tau sites and neuronal dysfunction. In the entire cohort, CSF p-tau181/Aβ40lumi and Aβ42/Aβ40lumi and mass spectrometry measurements for CSF pT217/T217, pT111/T111, pT231/T231 were the strongest predictors of amyloid PET status. Given their ability to denote individuals on an Alzheimer's disease pathological trajectory, these same markers (CSF pT217/T217, pT111/T111, p-tau/Aβ40lumi and t-tau/Aβ40lumi) were largely the best predictors of worse cognition in the entire cohort. When restricting analyses to amyloid-positive individuals, the strongest predictors of impaired cognition were tau PET, CSF t-tau/Aβ40lumi, p-tau181/Aβ40lumi, CSF pT217/217 and pT205/T205. Non-specific CSF measures of neuronal dysfunction and inflammation were poor predictors of amyloid PET and cognitive status. The current work utilized machine learning to understand the interrelationship structure and utility of a large number of biomarkers. The results demonstrate that, although the number of biomarkers has rapidly expanded, many are interrelated and few strongly predict clinical outcomes. Examining the entire corpus of available biomarkers simultaneously provides a meaningful framework to understand Alzheimer's disease pathobiological change as well as insight into which biomarkers may be most useful in Alzheimer's disease clinical practice and trials.
Collapse
Affiliation(s)
- Karin L Meeker
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Patrick H Luckett
- Department of Neurosurgery, Washington University in St Louis, St Louis, MO 63110, USA
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Diana A Hobbs
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Charles Chen
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - James Bollinger
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Shaney Flores
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Sarah Keefe
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Rachel L Henson
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Elizabeth M Herries
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Jason J Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
6
|
Lojo-Ramírez JA, Guerra-Gómez M, Marín-Cabañas AM, Fernández-Rodríguez P, Bernal Sánchez-Arjona M, Franco-Macías E, García-Solís D. Correlation Between Amyloid PET Imaging and Discordant Cerebrospinal Fluid Biomarkers Results in Patients with Suspected Alzheimer's Disease. J Alzheimers Dis 2024; 97:447-458. [PMID: 38143353 DOI: 10.3233/jad-230744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
BACKGROUND Although the concordance between cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers and amyloid-PET findings is well known, there are no data regarding the concordance of amyloid-PET with inconclusive CSF values of amyloid-β (Aβ)1 - 42 and p-tau for the diagnosis of AD. OBJECTIVE To investigate the relationship between the amyloid-PET results with discordant AD biomarkers values in CSF (Aβ1 - 42+/p-tau-or Aβ1 - 42-/p-tau+). METHODS An observational retrospective study, including 62 patients with mild cognitive impairment (32/62) or dementia (30/62), suspicious of AD who had undergone a lumbar puncture to determine CSF AD biomarkers, and presented discordant values in CSF between Aβ1 - 42 and p-tau (Aβ1 - 42+/p-tau-or Aβ1 - 42-/p-tau+). All of them, underwent an amyloid-PET with 18F-Florbetaben. An extensive neuropsychological testing as part of their diagnostic process (MMSE and TMA-93), was performed, and it was also obtained the Global Deterioration Scale. RESULTS Comparing the discordant CSF results of each patient with the cerebral amyloid-PET results, we found that in the group with Aβ1 - 42+ and p-tau-CSF values, the amyloid-PET was positive in 51.2% and negative in 48.8% of patients, while in the group with Aβ1 - 42-and p-Tau+ CSF values, the amyloid-PET was positive in 52.6% of patients and negative in 47.4% of them. No significant association was found (p = 0.951) between the results of amyloid-PET and the two divergent groups in CSF. CONCLUSIONS No significant relationship was observed between the results of discordant AD biomarkers in CSF and the result of amyloid-PET. No trend in amyloid-PET results was observed in relation to CSF biomarker values.
Collapse
Affiliation(s)
| | - Miriam Guerra-Gómez
- Department of Nuclear Medicine, Virgen del Rocío University Hospital, Seville, Spain
| | | | | | | | - Emilio Franco-Macías
- Memory Unit, Department of Neurology, Virgen del Rocío University Hospital, Seville, Spain
| | - David García-Solís
- Department of Nuclear Medicine, Virgen del Rocío University Hospital, Seville, Spain
| |
Collapse
|
7
|
Burnham SC, Iaccarino L, Pontecorvo MJ, Fleisher AS, Lu M, Collins EC, Devous MD. A review of the flortaucipir literature for positron emission tomography imaging of tau neurofibrillary tangles. Brain Commun 2023; 6:fcad305. [PMID: 38187878 PMCID: PMC10768888 DOI: 10.1093/braincomms/fcad305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease is defined by the presence of β-amyloid plaques and neurofibrillary tau tangles potentially preceding clinical symptoms by many years. Previously only detectable post-mortem, these pathological hallmarks are now identifiable using biomarkers, permitting an in vivo definitive diagnosis of Alzheimer's disease. 18F-flortaucipir (previously known as 18F-T807; 18F-AV-1451) was the first tau positron emission tomography tracer to be introduced and is the only Food and Drug Administration-approved tau positron emission tomography tracer (Tauvid™). It has been widely adopted and validated in a number of independent research and clinical settings. In this review, we present an overview of the published literature on flortaucipir for positron emission tomography imaging of neurofibrillary tau tangles. We considered all accessible peer-reviewed literature pertaining to flortaucipir through 30 April 2022. We found 474 relevant peer-reviewed publications, which were organized into the following categories based on their primary focus: typical Alzheimer's disease, mild cognitive impairment and pre-symptomatic populations; atypical Alzheimer's disease; non-Alzheimer's disease neurodegenerative conditions; head-to-head comparisons with other Tau positron emission tomography tracers; and technical considerations. The available flortaucipir literature provides substantial evidence for the use of this positron emission tomography tracer in assessing neurofibrillary tau tangles in Alzheimer's disease and limited support for its use in other neurodegenerative disorders. Visual interpretation and quantitation approaches, although heterogeneous, mostly converge and demonstrate the high diagnostic and prognostic value of flortaucipir in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Ming Lu
- Avid, Eli Lilly and Company, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
8
|
Lepinay E, Cicchetti F. Tau: a biomarker of Huntington's disease. Mol Psychiatry 2023; 28:4070-4083. [PMID: 37749233 DOI: 10.1038/s41380-023-02230-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023]
Abstract
Developing effective treatments for patients with Huntington's disease (HD)-a neurodegenerative disorder characterized by severe cognitive, motor and psychiatric impairments-is proving extremely challenging. While the monogenic nature of this condition enables to identify individuals at risk, robust biomarkers would still be extremely valuable to help diagnose disease onset and progression, and especially to confirm treatment efficacy. If measurements of cerebrospinal fluid neurofilament levels, for example, have demonstrated use in recent clinical trials, other proteins may prove equal, if not greater, relevance as biomarkers. In fact, proteins such as tau could specifically be used to detect/predict cognitive affectations. We have herein reviewed the literature pertaining to the association between tau levels and cognitive states, zooming in on Alzheimer's disease, Parkinson's disease and traumatic brain injury in which imaging, cerebrospinal fluid, and blood samples have been interrogated or used to unveil a strong association between tau and cognition. Collectively, these areas of research have accrued compelling evidence to suggest tau-related measurements as both diagnostic and prognostic tools for clinical practice. The abundance of information retrieved in this niche of study has laid the groundwork for further understanding whether tau-related biomarkers may be applied to HD and guide future investigations to better understand and treat this disease.
Collapse
Affiliation(s)
- Eva Lepinay
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
9
|
Quattrini G, Ferrari C, Pievani M, Geviti A, Ribaldi F, Scheffler M, Frisoni GB, Garibotto V, Marizzoni M. Unsupervised [ 18F]Flortaucipir cutoffs for tau positivity and staging in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2023; 50:3265-3275. [PMID: 37272955 PMCID: PMC10542510 DOI: 10.1007/s00259-023-06280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023]
Abstract
PURPOSE Several [18F]Flortaucipir cutoffs have been proposed for tau PET positivity (T+) in Alzheimer's disease (AD), but none were data-driven. The aim of this study was to establish and validate unsupervised T+ cutoffs by applying Gaussian mixture models (GMM). METHODS Amyloid negative (A-) cognitively normal (CN) and amyloid positive (A+) AD-related dementia (ADRD) subjects from ADNI (n=269) were included. ADNI (n=475) and Geneva Memory Clinic (GMC) cohorts (n=98) were used for validation. GMM-based cutoffs were extracted for the temporal meta-ROI, and validated against previously published cutoffs and visual rating. RESULTS GMM-based cutoffs classified less subjects as T+, mainly in the A- CN (<3.4% vs >28.5%) and A+ CN (<14.5% vs >42.9%) groups and showed higher agreement with visual rating (ICC=0.91 vs ICC<0.62) than published cutoffs. CONCLUSION We provided reliable data-driven [18F]Flortaucipir cutoffs for in vivo T+ detection in AD. These cutoffs might be useful to select participants in clinical and research studies.
Collapse
Affiliation(s)
- Giulia Quattrini
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Clarissa Ferrari
- FONDAZIONE POLIAMBULANZA ISTITUTO OSPEDALIERO via Bissolati, 57, 25124, Brescia, Italy
- Unit of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Andrea Geviti
- Unit of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Federica Ribaldi
- LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, 1205, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, 1205, Geneva, Switzerland
| | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - Giovanni B Frisoni
- LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, 1205, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, 1205, Geneva, Switzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocentre, Faculty of Medicine, University of Geneva, 1205, Geneva, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, 1205, Geneva, Switzerland
- Centre for Biomedical Imaging (CIBM), 1205, Geneva, Switzerland
| | - Moira Marizzoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy.
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy.
| |
Collapse
|
10
|
Wisch JK, Butt OH, Gordon BA, Schindler SE, Fagan AM, Henson RL, Yang C, Boerwinkle AH, Benzinger TLS, Holtzman DM, Morris JC, Cruchaga C, Ances BM. Proteomic clusters underlie heterogeneity in preclinical Alzheimer's disease progression. Brain 2023; 146:2944-2956. [PMID: 36542469 PMCID: PMC10316757 DOI: 10.1093/brain/awac484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Heterogeneity in progression to Alzheimer's disease (AD) poses challenges for both clinical prognosis and clinical trial implementation. Multiple AD-related subtypes have previously been identified, suggesting differences in receptivity to drug interventions. We identified early differences in preclinical AD biomarkers, assessed patterns for developing preclinical AD across the amyloid-tau-(neurodegeneration) [AT(N)] framework, and considered potential sources of difference by analysing the CSF proteome. Participants (n = 10) enrolled in longitudinal studies at the Knight Alzheimer Disease Research Center completed four or more lumbar punctures. These individuals were cognitively normal at baseline. Cerebrospinal fluid measures of amyloid-β (Aβ)42, phosphorylated tau (pTau181), and neurofilament light chain (NfL) as well as proteomics values were evaluated. Imaging biomarkers, including PET amyloid and tau, and structural MRI, were repeatedly obtained when available. Individuals were staged according to the amyloid-tau-(neurodegeneration) framework. Growth mixture modelling, an unsupervised clustering technique, identified three patterns of biomarker progression as measured by CSF pTau181 and Aβ42. Two groups (AD Biomarker Positive and Intermediate AD Biomarker) showed distinct progression from normal biomarker status to having biomarkers consistent with preclinical AD. A third group (AD Biomarker Negative) did not develop abnormal AD biomarkers over time. Participants grouped by CSF trajectories were re-classified using only proteomic profiles (AUCAD Biomarker Positive versus AD Biomarker Negative = 0.857, AUCAD Biomarker Positive versus Intermediate AD Biomarkers = 0.525, AUCIntermediate AD Biomarkers versus AD Biomarker Negative = 0.952). We highlight heterogeneity in the development of AD biomarkers in cognitively normal individuals. We identified some individuals who became amyloid positive before the age of 50 years. A second group, Intermediate AD Biomarkers, developed elevated CSF ptau181 significantly before becoming amyloid positive. A third group were AD Biomarker Negative over repeated testing. Our results could influence the selection of participants for specific treatments (e.g. amyloid-reducing versus other agents) in clinical trials. CSF proteome analysis highlighted additional non-AT(N) biomarkers for potential therapies, including blood-brain barrier-, vascular-, immune-, and neuroinflammatory-related targets.
Collapse
Affiliation(s)
- Julie K Wisch
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Omar H Butt
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L Henson
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anna H Boerwinkle
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Hope Center, Washington University in Saint Louis, St. Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
11
|
Barthélemy NR, Saef B, Li Y, Gordon BA, He Y, Horie K, Stomrud E, Salvadó G, Janelidze S, Sato C, Ovod V, Henson RL, Fagan AM, Benzinger TLS, Xiong C, Morris JC, Hansson O, Bateman RJ, Schindler SE. CSF tau phosphorylation occupancies at T217 and T205 represent improved biomarkers of amyloid and tau pathology in Alzheimer's disease. NATURE AGING 2023; 3:391-401. [PMID: 37117788 PMCID: PMC10154225 DOI: 10.1038/s43587-023-00380-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/03/2023] [Indexed: 04/30/2023]
Abstract
Cerebrospinal fluid (CSF) amyloid-β peptide (Aβ)42/Aβ40 and the concentration of tau phosphorylated at site 181 (p-tau181) are well-established biomarkers of Alzheimer's disease (AD). The present study used mass spectrometry to measure concentrations of nine phosphorylated and five nonphosphorylated tau species and phosphorylation occupancies (percentage phosphorylated/nonphosphorylated) at ten sites. In the present study we show that, in 750 individuals with a median age of 71.2 years, CSF pT217/T217 predicted the presence of brain amyloid by positron emission tomography (PET) slightly better than Aβ42/Aβ40 (P = 0.02). Furthermore, for individuals with positive brain amyloid by PET (n = 263), CSF pT217/T217 was more strongly correlated with the amount of amyloid (Spearman's ρ = 0.69) than Aβ42/Aβ40 (ρ = -0.42, P < 0.0001). In two independent cohorts of participants with symptoms of AD dementia (n = 55 and n = 90), CSF pT217/T217 and pT205/T205 were better correlated with tau PET measures than CSF p-tau181 concentration. These findings suggest that CSF pT217/T217 and pT205/T205 represent improved CSF biomarkers of amyloid and tau pathology in AD.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA.
| | - Benjamin Saef
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Rachel L Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
12
|
Mohammadi Z, Alizadeh H, Marton J, Cumming P. The Sensitivity of Tau Tracers for the Discrimination of Alzheimer's Disease Patients and Healthy Controls by PET. Biomolecules 2023; 13:290. [PMID: 36830659 PMCID: PMC9953528 DOI: 10.3390/biom13020290] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Hyperphosphorylated tau aggregates, also known as neurofibrillary tangles, are a hallmark neuropathological feature of Alzheimer's disease (AD). Molecular imaging of tau by positron emission tomography (PET) began with the development of [18F]FDDNP, an amyloid β tracer with off-target binding to tau, which obtained regional specificity through the differing distributions of amyloid β and tau in AD brains. A concerted search for more selective and affine tau PET tracers yielded compounds belonging to at least eight structural categories; 18F-flortaucipir, known variously as [18F]-T807, AV-1451, and Tauvid®, emerged as the first tau tracer approved by the American Food and Drug Administration. The various tau tracers differ concerning their selectivity over amyloid β, off-target binding at sites such as monoamine oxidase and neuromelanin, and degree of uptake in white matter. While there have been many reviews of molecular imaging of tau in AD and other conditions, there has been no systematic comparison of the fitness of the various tracers for discriminating between AD patient and healthy control (HC) groups. In this narrative review, we endeavored to compare the binding properties of the various tau tracers in vitro and the effect size (Cohen's d) for the contrast by PET between AD patients and age-matched HC groups. The available tracers all gave good discrimination, with Cohen's d generally in the range of two-three in culprit brain regions. Overall, Cohen's d was higher for AD patient groups with more severe illness. Second-generation tracers, while superior concerning off-target binding, do not have conspicuously higher sensitivity for the discrimination of AD and HC groups. We suppose that available pharmacophores may have converged on a maximal affinity for tau fibrils, which may limit the specific signal imparted in PET studies.
Collapse
Affiliation(s)
- Zohreh Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hadi Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Straße 10-14, D-01454 Radeberg, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
13
|
Dincer A, Chen CD, McKay NS, Koenig LN, McCullough A, Flores S, Keefe SJ, Schultz SA, Feldman RL, Joseph-Mathurin N, Hornbeck RC, Cruchaga C, Schindler SE, Holtzman DM, Morris JC, Fagan AM, Benzinger TLS, Gordon BA. APOE ε4 genotype, amyloid-β, and sex interact to predict tau in regions of high APOE mRNA expression. Sci Transl Med 2022; 14:eabl7646. [PMID: 36383681 PMCID: PMC9912474 DOI: 10.1126/scitranslmed.abl7646] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele is strongly linked with cerebral β-amyloidosis, but its relationship with tauopathy is less established. We investigated the relationship between APOE ε4 carrier status, regional amyloid-β (Aβ), magnetic resonance imaging (MRI) volumetrics, tau positron emission tomography (PET), APOE messenger RNA (mRNA) expression maps, and cerebrospinal fluid phosphorylated tau (CSF ptau181). Three hundred fifty participants underwent imaging, and 270 had ptau181. We used computational models to evaluate the main effect of APOE ε4 carrier status on regional neuroimaging values and then the interaction of ε4 status and global Aβ on regional tau PET and brain volumes as well as CSF ptau181. Separately, we also examined the additional interactive influence of sex. We found that, for the same degree of Aβ burden, APOE ε4 carriers showed greater tau PET signal relative to noncarriers in temporal regions, but no interaction was present for MRI volumes or CSF ptau181. This potentiation of tau aggregation irrespective of sex occurred in brain regions with high APOE mRNA expression, suggesting local vulnerabilities to tauopathy. There were greater effects of APOE genotype in females, although the interactive sex effects did not strongly mirror mRNA expression. Pathology is not homogeneously expressed throughout the brain but mirrors underlying biological patterns such as gene expression.
Collapse
Affiliation(s)
- Aylin Dincer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lauren N Koenig
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah J Keefe
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephanie A Schultz
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rebecca L Feldman
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Suzanne E Schindler
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - David M Holtzman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammie LS Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychological & Brain Sciences, Washington University, Saint Louis, MO, USA
| |
Collapse
|
14
|
Kim Y, Park H, Kim Y, Kim SH, Lee JH, Yang H, Kim SJ, Li CM, Lee H, Na DH, Moon S, Shin Y, Kam TI, Lee HW, Kim S, Song JJ, Jung YK. Pathogenic Role of RAGE in Tau Transmission and Memory Deficits. Biol Psychiatry 2022; 93:829-841. [PMID: 36759256 DOI: 10.1016/j.biopsych.2022.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND In tauopathies, brain regions with tau accumulation strongly correlate with clinical symptoms, and spreading of misfolded tau along neural network leads to disease progression. However, the underlying mechanisms by which tau proteins enter neurons during pathological propagation remain unclear. METHODS To identify membrane receptors responsible for neuronal propagation of tau oligomers, we established a cell-based tau uptake assay and screened complementary DNA expression library. Tau uptake and propagation were analyzed in vitro and in vivo using a microfluidic device and stereotactic injection. The cognitive function of mice was assessed using behavioral tests. RESULTS From a genome-wide cell-based functional screening, RAGE (receptor for advanced glycation end products) was isolated to stimulate the cellular uptake of tau oligomers. Rage deficiency reduced neuronal uptake of pathological tau prepared from rTg4510 mouse brains or cerebrospinal fluid from patients with Alzheimer's disease and slowed tau propagation between neurons cultured in a 3-chamber microfluidic device. RAGE levels were increased in the brains of rTg4510 mice and tau oligomer-treated neurons. Rage knockout decreased tau transmission in the brains of nontransgenic mice after injection with Alzheimer's disease patient-derived tau and ameliorated memory loss after injection with GFP-P301L-tau-AAV. Treatment of RAGE antagonist FPS-ZM1 blocked transsynaptic tau propagation and inflammatory responses and alleviated cognitive impairment in rTg4510 mice. CONCLUSIONS These results suggest that in neurons and microglia, RAGE binds to pathological tau and facilitates neuronal tau pathology progression and behavioral deficits in tauopathies.
Collapse
Affiliation(s)
- Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Republic of Korea; School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Park
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Youngwon Kim
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seo-Hyun Kim
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Hoon Lee
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Hanseul Yang
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seo Jin Kim
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cathena Meiling Li
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Haneul Lee
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Do-Hyeong Na
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seowon Moon
- School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yumi Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Tae-In Kam
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Han-Woong Lee
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Ji-Joon Song
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yong-Keun Jung
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Republic of Korea; School of the Biological Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Strikwerda-Brown C, Hobbs DA, Gonneaud J, St-Onge F, Binette AP, Ozlen H, Provost K, Soucy JP, Buckley RF, Benzinger TLS, Morris JC, Villemagne VL, Doré V, Sperling RA, Johnson KA, Rowe CC, Gordon BA, Poirier J, Breitner JCS, Villeneuve S. Association of Elevated Amyloid and Tau Positron Emission Tomography Signal With Near-Term Development of Alzheimer Disease Symptoms in Older Adults Without Cognitive Impairment. JAMA Neurol 2022; 79:975-985. [PMID: 35907254 PMCID: PMC9339146 DOI: 10.1001/jamaneurol.2022.2379] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/24/2022] [Indexed: 12/12/2022]
Abstract
Importance National Institute on Aging-Alzheimer's Association (NIA-AA) workgroups have proposed biological research criteria intended to identify individuals with preclinical Alzheimer disease (AD). Objective To assess the clinical value of these biological criteria to identify older individuals without cognitive impairment who are at near-term risk of developing symptomatic AD. Design, Setting, and Participants This longitudinal cohort study used data from 4 independent population-based cohorts (PREVENT-AD, HABS, AIBL, and Knight ADRC) collected between 2003 and 2021. Participants were older adults without cognitive impairment with 1 year or more of clinical observation after amyloid β and tau positron emission tomography (PET). Median clinical follow-up after PET ranged from 1.94 to 3.66 years. Exposures Based on binary assessment of global amyloid burden (A) and a composite temporal region of tau PET uptake (T), participants were stratified into 4 groups (A+T+, A+T-, A-T+, A-T-). Presence (+) or absence (-) of neurodegeneration (N) was assessed using temporal cortical thickness. Main Outcomes and Measures Each cohort was analyzed separately. Primary outcome was clinical progression to mild cognitive impairment (MCI), identified by a Clinical Dementia Rating score of 0.5 or greater in Knight ADRC and by consensus committee review in the other cohorts. Clinical raters were blind to imaging, genetic, and fluid biomarker data. A secondary outcome was cognitive decline, based on a slope greater than 1.5 SD below the mean of an independent subsample of individuals without cognitive impairment. Outcomes were compared across the biomarker groups. Results Among 580 participants (PREVENT-AD, 128; HABS, 153; AIBL, 48; Knight ADRC, 251), mean (SD) age ranged from 67 (5) to 76 (6) years across cohorts, with between 55% (137/251) and 74% (95/128) female participants. Across cohorts, 33% to 83% of A+T+ participants progressed to MCI during follow-up (mean progression time, 2-2.72 years), compared with less than 20% of participants in other biomarker groups. Progression further increased to 43% to 100% when restricted to A+T+(N+) individuals. Cox proportional hazard ratios for progression to MCI in the A+T+ group vs other biomarker groups were all 5 or greater. Many A+T+ nonprogressors also showed longitudinal cognitive decline, while cognitive trajectories in other groups remained predominantly stable. Conclusions and Relevance The clinical prognostic value of NIA-AA research criteria was confirmed in 4 independent cohorts, with most A+T+(N+) older individuals without cognitive impairment developing AD symptoms within 2 to 3 years.
Collapse
Affiliation(s)
- Cherie Strikwerda-Brown
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Diana A. Hobbs
- Washington University School of Medicine, St Louis, Missouri
| | - Julie Gonneaud
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Inserm, Inserm UMR-S U1237, Université de Caen-Normandie, GIP Cyceron, Caen, France
| | - Frédéric St-Onge
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Alexa Pichet Binette
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Hazal Ozlen
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Karine Provost
- Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Paul Soucy
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Rachel F. Buckley
- Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Brigham and Women’s Hospital, Boston, Massachusetts
- Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | | | - John C. Morris
- Washington University School of Medicine, St Louis, Missouri
| | | | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia
| | - Reisa A. Sperling
- Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Keith A. Johnson
- Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Christopher C. Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia
| | - Brian A. Gordon
- Washington University School of Medicine, St Louis, Missouri
| | - Judes Poirier
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - John C. S. Breitner
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Cogswell PM, Wiste HJ, Mielke MM, Schwarz CG, Weigand SD, Lowe VJ, Therneau TM, Knopman DS, Graff-Radford J, Vemuri P, Senjem ML, Gunter JL, Algeciras-Schimnich A, Petersen RC, Jack CR. CSF phosphorylated tau as an indicator of subsequent tau accumulation. Neurobiol Aging 2022; 117:189-200. [PMID: 35764037 PMCID: PMC9361359 DOI: 10.1016/j.neurobiolaging.2022.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 11/19/2022]
Abstract
We evaluated the relationship between baseline CSF p-tau181 and the rate of tau PET change in the temporal meta-ROI and entorhinal cortex (ERC) and how it varied by amyloid level (CSF Aβ42 or amyloid PET) among 143 individuals from the Mayo Clinic Study of Aging and Mayo Alzheimer Disease Research Center. Higher CSF p-tau181, lower CSF Aβ42, and higher amyloid PET levels were associated with faster rates of tau PET change in both the temporal meta-ROI and ERC. In the temporal meta-ROI, longitudinal tau PET accumulation occurred primarily in participants with abnormal biomarker levels and a diagnosis of dementia, which supports the hypothesis that tau aggregation begins later in the disease process. Compared to the temporal meta-ROI, the ERC showed greater change in tau PET in non-demented participants but less change in later disease stages, supporting ERC as a more sensitive marker of early tau PET changes but with less dynamic range over the disease spectrum. We found both amyloid and CSF p-tau181 were associated with rates of tau PET change but there were some differences in associations by region, amyloid biomarker, and disease stage.
Collapse
Affiliation(s)
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Stephen D Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Terry M Therneau
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Information Technology, Mayo Clinic, Rochester, MN USA
| | | | | | - Ronald C Petersen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
17
|
Zhou J, Benoit M, Sharoar MG. Recent advances in pre-clinical diagnosis of Alzheimer's disease. Metab Brain Dis 2022; 37:1703-1725. [PMID: 33900524 DOI: 10.1007/s11011-021-00733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia with currently no known cures or disease modifying treatments (DMTs), despite much time and effort from the field. Diagnosis and intervention of AD during the early pre-symptomatic phase of the disease is thought to be a more effective strategy. Therefore, the detection of biomarkers has emerged as a critical tool for monitoring the effect of new AD therapies, as well as identifying patients most likely to respond to treatment. The establishment of the amyloid/tau/neurodegeneration (A/T/N) framework in 2018 has codified the contexts of use of AD biomarkers in neuroimaging and bodily fluids for research and diagnostic purposes. Furthermore, a renewed drive for novel AD biomarkers and innovative methods of detection has emerged with the goals of adding additional insight to disease progression and discovery of new therapeutic targets. The use of biomarkers has accelerated the development of AD drugs and will bring new therapies to patients in need. This review highlights recent methods utilized to diagnose antemortem AD.
Collapse
Affiliation(s)
- John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
- Molecular Medicine Program, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Marc Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA.
| |
Collapse
|
18
|
Therapeutic Potential of Different Natural Products for the Treatment of Alzheimer’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6873874. [PMID: 35910833 PMCID: PMC9337964 DOI: 10.1155/2022/6873874] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/09/2022] [Indexed: 12/03/2022]
Abstract
A high incidence of dementia (60–80%) and a high rate of memory loss are two of the most common symptoms of Alzheimer's disease (AD), which affects the elderly. Researchers have recommended that traditional Chinese medicine (TCM) and Indian medicines can be used to prevent and cure AD. Several studies have linked neuroinflammation linked to amyloid-β (Aβ) deposition in the brain to the pathophysiology of neurodegenerative disorders. As a result, more research is needed to determine the role of inflammation in neurodegeneration. Increased microglial activation, cytokine production, reactive oxygen species (ROS), and nuclear factor kappa B (NF-κB) all play a role in the inflammatory process of AD. This review focuses on the role of neuroinflammation in neuroprotection and the molecular processes used by diverse natural substances, phytochemicals, and herbal formulations in distinct signaling pathways. Currently, researchers are focusing on pharmacologically active natural compounds with the anti-neuroinflammatory potential, making them a possible contender for treating AD. Furthermore, the researchers investigated the limits of past studies on TCM, Indian Ayurveda, and AD. Numerous studies have been carried out to examine the effects of medicinal whole-plant extracts on AD. Clinical investigations have shown that lignans, flavonoids, tannins, polyphenols, triterpenoids, sterols, and alkaloids have anti-inflammatory, antiamyloidogenic, anticholinesterase, and antioxidant properties. This review summarizes information about numerous medicinal plants and isolated compounds used in the treatment of AD and a list of further references.
Collapse
|
19
|
Schindler SE. Predicting Symptom Onset in Sporadic Alzheimer's Disease: "How Long Do I Have?". J Alzheimers Dis 2022; 90:975-979. [PMID: 35213383 DOI: 10.3233/jad-215722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Predicting not just if but when cognitively normal individuals will develop the onset of Alzheimer's disease (AD) dementia seems increasingly feasible, as evidenced by converging findings from several approaches and cohorts. These estimates may improve the efficiency of clinical trials by better identifying cognitively normal individuals at high risk of developing AD symptoms. As models are refined, the implications of disclosing estimates of the age of AD symptom onset must be examined, since telling a cognitively normal individual the age they are expected to develop AD symptoms may have different implications than disclosing increased risk for AD dementia.
Collapse
Affiliation(s)
- Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
20
|
Mahaman YAR, Embaye KS, Huang F, Li L, Zhu F, Wang JZ, Liu R, Feng J, Wang X. Biomarkers used in Alzheimer's disease diagnosis, treatment, and prevention. Ageing Res Rev 2022; 74:101544. [PMID: 34933129 DOI: 10.1016/j.arr.2021.101544] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), being the number one in terms of dementia burden, is an insidious age-related neurodegenerative disease and is presently considered a global public health threat. Its main histological hallmarks are the Aβ senile plaques and the P-tau neurofibrillary tangles, while clinically it is marked by a progressive cognitive decline that reflects the underlying synaptic loss and neurodegeneration. Many of the drug therapies targeting the two pathological hallmarks namely Aβ and P-tau have been proven futile. This is probably attributed to the initiation of therapy at a stage where cognitive alterations are already obvious. In other words, the underlying neuropathological changes are at a stage where these drugs lack any therapeutic value in reversing the damage. Therefore, there is an urgent need to start treatment in the very early stage where these changes can be reversed, and hence, early diagnosis is of primordial importance. To this aim, the use of robust and informative biomarkers that could provide accurate diagnosis preferably at an earlier phase of the disease is of the essence. To date, several biomarkers have been established that, to a different extent, allow researchers and clinicians to evaluate, diagnose, and more specially exclude other related pathologies. In this study, we extensively reviewed data on the currently explored biomarkers in terms of AD pathology-specific and non-specific biomarkers and highlighted the recent developments in the diagnostic and theragnostic domains. In the end, we have presented a separate elaboration on aspects of future perspectives and concluding remarks.
Collapse
|
21
|
Bridel C, Somers C, Sieben A, Rozemuller A, Niemantsverdriet E, Struyfs H, Vermeiren Y, Van Broeckhoven C, De Deyn PP, Bjerke M, Nagels G, Teunissen CE, Engelborghs S. Associating Alzheimer’s disease pathology with its cerebrospinal fluid biomarkers. Brain 2022; 145:4056-4064. [DOI: 10.1093/brain/awac013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 11/12/2022] Open
Abstract
Abstract
Alzheimer’s disease cerebrospinal fluid (CSF) biomarkers 42 amino acid long amyloid-β peptide (Aβ1-42), total tau protein (T-tau), and tau protein phosphorylated at threonine 181 (P-tau181) are considered surrogate biomarkers of Alzheimer’s disease pathology, and significantly improve diagnostic accuracy. Their ability to reflect neuropathological changes later in the disease course is not well characterized. This study aimed to assess the potential of CSF biomarkers measured in mid- to late-stage Alzheimer’s disease to reflect post mortem neuropathological changes. Individuals were selected from 2 autopsy cohorts of Alzheimer’s disease patients in Antwerp and Amsterdam. Neuropathological diagnosis was performed according to the updated consensus National Institute on Aging-Alzheimer’s Association guidelines by Montine et al, which includes quantification of amyloid beta plaque, neurofibrillary tangle, and neuritic plaque load. CSF samples were analyzed for Aβ1-42, T-tau, and P-tau181 by ELISA. 114 cases of pure definite Alzheimer’s disease were included in the study (mean age 74 years, disease duration 6 years at CSF sampling, 50% females). Median interval between CSF sampling and death was one year. We found no association between Aβ1-42 and Alzheimer’s disease neuropathological change profile. In contrast, an association of P-tau181 and T-tau with Alzheimer’s disease neuropathological change profile was observed. P-tau181 was associated with all three individual Montine scores, and the associations became stronger and more significant as the interval between lumbar puncture and death increased. T-tau was also associated with all three Montine scores, but in individuals with longer intervals from lumbar puncture to death only. Stratification of the cohort according to APOE ε4 carrier status revealed that the associations applied mostly to APOE ε4 non-carriers. Our data suggest that similarly to what has been reported for Aβ1-42, plateau levels of P-tau181 and T-tau are reached during the disease course, albeit at later disease stages, reducing the potential of tau biomarkers to monitor Alzheimer’s disease pathology as the disease progresses. As a consequence, CSF biomarkers, which are performant for clinical diagnosis of early Alzheimer’s disease, may not be well suited for staging or monitoring Alzheimer’s disease pathology as it progresses through later stages.
Collapse
Affiliation(s)
- Claire Bridel
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, The Netherlands
- Department of Clinical Neurosciences, Division of Neurology, Geneva University Hospital, Geneva, Switzerland
| | - Charisse Somers
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Anne Sieben
- Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Annemieke Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, The Netherlands
| | - Ellis Niemantsverdriet
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Hanne Struyfs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Yannick Vermeiren
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Chair Group of Nutritional Biology, Division of Human Nutrition and Health, Wageningen University and Research (WUR), Wageningen, The Netherlands
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium
- Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Peter P. De Deyn
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Brussels, Belgium
- Universitair Ziekenhuis Brussel, Laboratory of Neurochemistry, Brussels, Belgium
| | - Guy Nagels
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Brussels, Belgium
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, The Netherlands
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Brussels, Belgium
| |
Collapse
|
22
|
Paczynski MM, Day GS. Alzheimer Disease Biomarkers in Clinical Practice: A Blood-Based Diagnostic Revolution. J Prim Care Community Health 2022; 13:21501319221141178. [PMID: 36475976 PMCID: PMC9742698 DOI: 10.1177/21501319221141178] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022] Open
Abstract
An estimated 6.1 million Americans live with cognitive impairment-a number that is expected to triple by 2050. Alzheimer disease (AD) is the most common cause of impairment. The development of blood-based biomarkers capable of detecting pathological changes of AD in living patients has the potential to revolutionize the diagnostic approach to cognitive impairment by enabling screening for AD using accessible, non-invasive measures of amyloid and tau neuropathology, with accuracy that increasingly approaches that seen with "gold standard" positron emission tomography and cerebrospinal fluid measures. Demand for biomarker testing is expected to intensify with the emergence of effective treatments for AD and related dementias. Clinicians in all fields must prepare to meet this demand. Primary care practitioners are well positioned to support dementia diagnosis and management, including the application and interpretation of biomarkers. This article reviews the current uses of AD biomarkers and the potential applications of emerging blood-based AD biomarkers in clinical practice.
Collapse
|
23
|
Altuna-Azkargorta M, Mendioroz-Iriarte M. Blood biomarkers in Alzheimer's disease. NEUROLOGÍA (ENGLISH EDITION) 2021; 36:704-710. [PMID: 34752348 DOI: 10.1016/j.nrleng.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/01/2018] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Early diagnosis of Alzheimer disease (AD) through the use of biomarkers could assist in the implementation and monitoring of early therapeutic interventions, and has the potential to significantly modify the course of the disease. DEVELOPMENT The classic cerebrospinal fluid and approved structural and functional neuroimaging biomarkers are of limited clinical application given their invasive nature and/or high cost. The identification of more accessible and less costly biomarkers, such as blood biomarkers, would increase their use in clinical practice. We review the available published evidence on the main blood biochemical biomarkers potentially useful for diagnosing AD. CONCLUSIONS Blood biomarkers are more cost- and time-effective than CSF biomarkers. However, immediate applicability in clinical practice is relatively unlikely. The main limitations come from the difficulty of measuring and standardising thresholds between different laboratories and the failure to replicate results. Of all the molecules studied, apoptosis and neurodegeneration biomarkers and the biomarker panels obtained through "omics" approaches, such as isolated or combined metabolomics, offer the most promising results.
Collapse
Affiliation(s)
- M Altuna-Azkargorta
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.
| | - M Mendioroz-Iriarte
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, Spain
| |
Collapse
|
24
|
Pelgrim TA, Beran M, Twait EL, Geerlings MI, Vonk JM. Cross-sectional associations of tau protein biomarkers with semantic and episodic memory in older adults without dementia: A systematic review and meta-analysis. Ageing Res Rev 2021; 71:101449. [PMID: 34400308 DOI: 10.1016/j.arr.2021.101449] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
Pathological tau is suggested to play a role in cognitive deterioration in the preclinical phase of Alzheimer's disease. We investigated cross-sectional associations of tau burden with episodic and semantic memory performance in older adults without dementia. A systematic search in MEDLINE (via PubMed), PsychINFO, and Embase resulted in 24 eligible studies for meta-analysis. Tau burden was assessed using CSF, PET, or histopathological measures. All studies evaluated associations of tau with episodic memory: weighted effect sizes were -0.46 (95 % CI [-0.73; -0.20], p < .001) for episodic composite scores, -0.19 ([-0.36; -0.03], p = .024) for delayed word list recall, and -0.05 ([-0.14; 0.04], p = .257) for logical memory. Fourteen studies evaluated associations of tau with semantic memory: weighted effect sizes were -0.28 ([-0.52; -0.04], p = .023) for semantic composite scores, -0.06 ([-0.16; 0.03], p = .194) for semantic fluency, and 0.06 ([-0.06; 0.18], p = .319) for picture naming. Our findings indicate that tau burden related to both episodic and semantic memory impairment in older individuals without a diagnosis of mild cognitive impairment or manifest dementia, with episodic composite scores showing the strongest association with tau burden. Future potential lies in developing more sensitive scores to detect this subtle cognitive impairment, which could contribute to early identification of individuals in the preclinical phase of Alzheimer's disease, thereby improving early diagnosis and timely intervention.
Collapse
|
25
|
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
|
26
|
Okafor M, Nye JA, Shokouhi M, Shaw LM, Goldstein F, Hajjar I. 18F-Flortaucipir PET Associations with Cerebrospinal Fluid, Cognition, and Neuroimaging in Mild Cognitive Impairment due to Alzheimer's Disease. J Alzheimers Dis 2021; 74:589-601. [PMID: 32065800 DOI: 10.3233/jad-191330] [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: 01/01/2023]
Abstract
BACKGROUND Tau positron emission tomography (PET) imaging is used in research, but its relation to cerebrospinal fluid (CSF) tau and other Alzheimer's disease (AD)-related clinical measures is unclear in mild cognitive impairment with AD biomarkers (MCI-AD). OBJECTIVE To determine associations between 18F-flortaucipir PET and CSF AD biomarkers, cognitive functioning, and neuroimaging measures in MCI-AD. METHODS In 29 participants 50 years or older with MCI-AD, 18F-flortaucipir PET, CSF total tau (T-tau), phosphorylated tau181p (P-tau), amyloid-β (Aβ), structural MRI, and neuropsychological testing were collected as baseline assessments of an ongoing clinical trial. 11C-Pittsburgh compound B PET was simultaneously conducted in 20 participants. Associations between 18F-flortaucipir PET and these measures were assessed by multiple linear regression adjusted for potential confounders and using global, lobar, and voxel-wise standardized uptake value ratio (SUVr). RESULTS Whole brain 18F-flortaucipir SUVr was significantly associated with CSF T-tau (r = 0.68, p < 0.001) and P-tau (r = 0.42, p = 0.04) after adjusting for age, sex, race, and education, with strongest associations in the temporal region (T-tau: r = 0.69, p < 0.001; P-tau: r = 0.49, p = 0.02). Voxel-wise analysis confirmed these regional associations. 18F-flortaucipir PET was also associated with CSF Aβ (r = -0.45, p = 0.03), episodic memory (r = -0.61, p = 0.001), visuospatial working memory (r = -0.46, p = 0.02), and brain cortical thickness (r = -0.44, p = 0.03) but not hippocampal volume. In the amyloid PET subset, although 11C-PiB PET associated strongly with 18F-flortaucipir (r = 0.79, p≤0.001), associations were stronger between 11C-PiB and key outcomes, compared to 18F-flortaucipir. CONCLUSION 18F-flortaucipir PET is moderately associated with CSF AD biomarkers and other AD-related phenotypes. The associations in this MCI-AD sample are stronger than previously described in other populations.
Collapse
Affiliation(s)
- Maureen Okafor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathon A Nye
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Mahsa Shokouhi
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Felicia Goldstein
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ihab Hajjar
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.,Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
27
|
Wolters EE, Dodich A, Boccardi M, Corre J, Drzezga A, Hansson O, Nordberg A, Frisoni GB, Garibotto V, Ossenkoppele R. Clinical validity of increased cortical uptake of [ 18F]flortaucipir on PET as a biomarker for Alzheimer's disease in the context of a structured 5-phase biomarker development framework. Eur J Nucl Med Mol Imaging 2021; 48:2097-2109. [PMID: 33547556 PMCID: PMC8175307 DOI: 10.1007/s00259-020-05118-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE In 2017, the Geneva Alzheimer's disease (AD) Biomarker Roadmap initiative adapted the framework of the systematic validation of oncological diagnostic biomarkers to AD biomarkers, with the aim to accelerate their development and implementation in clinical practice. With this work, we assess the maturity of [18F]flortaucipir PET and define its research priorities. METHODS The level of maturity of [18F]flortaucipir was assessed based on the AD Biomarker Roadmap. The framework assesses analytical validity (phases 1-2), clinical validity (phases 3-4), and clinical utility (phase 5). RESULTS The main aims of phases 1 (rationale for use) and 2 (discriminative ability) have been achieved. [18F]Flortaucipir binds with high affinity to paired helical filaments of tau and has favorable kinetic properties and excellent discriminative accuracy for AD. The majority of secondary aims of phase 2 were fully achieved. Multiple studies showed high correlations between ante-mortem [18F]flortaucipir PET and post-mortem tau (as assessed by histopathology), and also the effects of covariates on tracer binding are well studied. The aims of phase 3 (early detection ability) were only partially or preliminarily achieved, and the aims of phases 4 and 5 were not achieved. CONCLUSION Current literature provides partial evidence for clinical utility of [18F]flortaucipir PET. The aims for phases 1 and 2 were mostly achieved. Phase 3 studies are currently ongoing. Future studies including representative MCI populations and a focus on healthcare outcomes are required to establish full maturity of phases 4 and 5.
Collapse
Affiliation(s)
- E E Wolters
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - A Dodich
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Centre for Mind/Brain Sciences-CIMeC, University of Trento, Rovereto, Italy
| | - M Boccardi
- Late Translational Dementia Studies Group, German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Rostock, Germany
| | - J Corre
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- CURIC, Centre Universitaire Romand d'Implants Cochléaires, Department of Clinical Neurosciences, University of Geneva, Geneva, Switzerland
| | - A Drzezga
- Faculty of Medicine, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Molecular Organization of the Brain, Research Center Jülich, Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - O Hansson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - A Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - G B Frisoni
- LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
- Memory Clinic, University Hospital, Geneva, Switzerland
| | - V Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
| | - R Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Salvadó G, Grothe MJ, Groot C, Moscoso A, Schöll M, Gispert JD, Ossenkoppele R. Differential associations of APOE-ε2 and APOE-ε4 alleles with PET-measured amyloid-β and tau deposition in older individuals without dementia. Eur J Nucl Med Mol Imaging 2021; 48:2212-2224. [PMID: 33521872 PMCID: PMC8175302 DOI: 10.1007/s00259-021-05192-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/03/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE To examine associations between the APOE-ε2 and APOE-ε4 alleles and core Alzheimer's disease (AD) pathological hallmarks as measured by amyloid-β (Aβ) and tau PET in older individuals without dementia. METHODS We analyzed data from 462 ADNI participants without dementia who underwent Aβ ([18F]florbetapir or [18F]florbetaben) and tau ([18F]flortaucipir) PET, structural MRI, and cognitive testing. Employing APOE-ε3 homozygotes as the reference group, associations between APOE-ε2 and APOE-ε4 carriership with global Aβ PET and regional tau PET measures (entorhinal cortex (ERC), inferior temporal cortex, and Braak-V/VI neocortical composite regions) were investigated using linear regression models. In a subset of 156 participants, we also investigated associations between APOE genotype and regional tau accumulation over time using linear mixed models. Finally, we assessed whether Aβ mediated the cross-sectional and longitudinal associations between APOE genotype and tau. RESULTS Compared to APOE-ε3 homozygotes, APOE-ε2 carriers had lower global Aβ burden (βstd [95% confidence interval (CI)]: - 0.31 [- 0.45, - 0.16], p = 0.034) but did not differ on regional tau burden or tau accumulation over time. APOE-ε4 participants showed higher Aβ (βstd [95%CI]: 0.64 [0.42, 0.82], p < 0.001) and tau burden (βstd range: 0.27-0.51, all p < 0.006). In mediation analyses, APOE-ε4 only retained an Aβ-independent effect on tau in the ERC. APOE-ε4 showed a trend towards increased tau accumulation over time in Braak-V/VI compared to APOE-ε3 homozygotes (βstd [95%CI]: 0.10 [- 0.02, 0.18], p = 0.11), and this association was fully mediated by baseline Aβ. CONCLUSION Our data suggest that the established protective effect of the APOE-ε2 allele against developing clinical AD is primarily linked to resistance against Aβ deposition rather than tau pathology.
Collapse
Affiliation(s)
- Gemma Salvadó
- Alzheimer Prevention Program, Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30 08005, Barcelona, Spain.
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Michel J Grothe
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot, s/n 41013, Seville, Spain.
| | - Colin Groot
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alexis Moscoso
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - Juan Domingo Gispert
- Alzheimer Prevention Program, Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30 08005, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| |
Collapse
|
29
|
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
|
30
|
Rostamzadeh A, Jessen F. [Predictive Diagnosis of Alzheimer's Dementia]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2021; 89:254-266. [PMID: 34005829 DOI: 10.1055/a-1370-3142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Expanding technologies of early disease detection allow to identify Alzheimer's disease (AD) long before symptom onset. Hence, patients are increasingly demanding for these diagnostic procedures. Biomarker-based early detection of AD is therefore increasingly important in the clinical work-up. This article gives an overview of predictive procedures in the field of Alzheimer's dementia.
Collapse
|
31
|
Boerwinkle AH, Wisch JK, Chen CD, Gordon BA, Butt OH, Schindler SE, Sutphen C, Flores S, Dincer A, Benzinger TLS, Fagan AM, Morris JC, Ances BM. Temporal Correlation of CSF and Neuroimaging in the Amyloid-Tau-Neurodegeneration Model of Alzheimer Disease. Neurology 2021; 97:e76-e87. [PMID: 33931538 DOI: 10.1212/wnl.0000000000012123] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/23/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To evaluate temporal correlations between CSF and neuroimaging (PET and MRI) measures of amyloid, tau, and neurodegeneration in relation to Alzheimer disease (AD) progression. METHODS A total of 371 cognitively unimpaired and impaired participants enrolled in longitudinal studies of AD had both CSF (β-amyloid [Aβ]42, phosphorylated tau181, total tau, and neurofilament light chain) and neuroimaging (Pittsburgh compound B [PiB] PET, flortaucipir PET, and structural MRI) measures. The pairwise time interval between CSF and neuroimaging measures was binned into 2-year periods. Spearman correlations identified the time bin when CSF and neuroimaging measures most strongly correlated. CSF and neuroimaging measures were then binarized as biomarker-positive or biomarker-negative using Gaussian mixture modeling. Cohen kappa coefficient identified the time bin when CSF measures best agreed with corresponding neuroimaging measures when determining amyloid, tau, and neurodegeneration biomarker positivity. RESULTS CSF Aβ42 and PiB PET showed maximal correlation when collected within 6 years of each other (R ≈ -0.5). CSF phosphorylated tau181 and flortaucipir PET showed maximal correlation when CSF was collected 4 to 8 years prior to PET (R ≈ 0.4). CSF neurofilament light chain and cortical thickness showed low correlation, regardless of time interval (R avg ≈ -0.3). Similarly, CSF total tau and cortical thickness had low correlation, regardless of time interval (R avg < -0.2). CONCLUSIONS CSF Aβ42 and PiB PET best agree when acquired in close temporal proximity, whereas CSF phosphorylated tau precedes flortaucipir PET by 4 to 8 years. CSF and neuroimaging measures of neurodegeneration have low correspondence and are not interchangeable at any time interval.
Collapse
Affiliation(s)
- Anna H Boerwinkle
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Julie K Wisch
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Charles D Chen
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Brian A Gordon
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Omar H Butt
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Suzanne E Schindler
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Courtney Sutphen
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Shaney Flores
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Aylin Dincer
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Tammie L S Benzinger
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Anne M Fagan
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - John C Morris
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Beau M Ances
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO.
| |
Collapse
|
32
|
Use of Alzheimer's Disease Cerebrospinal Fluid Biomarkers in A Tertiary Care Memory Clinic. Can J Neurol Sci 2021; 49:203-209. [PMID: 33845924 DOI: 10.1017/cjn.2021.67] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers are promising tools to help identify the underlying pathology of neurocognitive disorders. In this manuscript, we report our experience with AD CSF biomarkers in 262 consecutive patients in a tertiary care memory clinic. METHODS We retrospectively reviewed 262 consecutive patients who underwent lumbar puncture (LP) and CSF measurement of AD biomarkers (Aβ1-42, total tau or t-tau, and p-tau181). We studied the safety of the procedure and its impact on patient's diagnosis and management. RESULTS The LP allowed to identify underlying AD pathology in 72 of the 121 patients (59%) with early onset amnestic mild cognitive impairment (aMCI) with a high probability of progression to AD; to distinguish the behavioral/dysexecutive variant of AD from the behavioral variant of frontotemporal dementia (bvFTD) in 25 of the 45 patients (55%) with an atypical neurobehavioral profile; to identify AD as the underlying pathology in 15 of the 27 patients (55%) with atypical or unclassifiable primary progressive aphasia (PPA); and to distinguish AD from other disorders in 9 of the 29 patients (31%) with psychiatric differential diagnoses and 19 of the 40 patients (47%) with lesional differential diagnoses (normal pressure hydrocephalus, encephalitis, prion disease, etc.). No major complications occurred following the LP. INTERPRETATION Our results suggest that CSF analysis is a safe and effective diagnostic tool in select patients with neurocognitive disorders. We advocate for a wider use of this biomarker in tertiary care memory clinics in Canada.
Collapse
|
33
|
Sun HH, Lin MY, Nouchi R, Wang PN, Cheng CH. Neuromagnetic evidence of abnormal automatic inhibitory function in subjective memory complaint. Eur J Neurosci 2021; 53:3350-3361. [PMID: 33754412 DOI: 10.1111/ejn.15196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 02/21/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Subjective memory complaint (SMC), a self-perceived worsening in memory capacity concurrent with normal performance on standardized cognitive assessments, is considered a risk factor for the development of Alzheimer's disease (AD). Deficient sensory gating (SG), referring to the lack of automatic inhibition of neural responses to the second identical stimulus, has been documented in prodromal and incident AD patients. However, it remains unknown whether the cognitively normal elderly with SMC demonstrate alterations of SG function compared with those without SMC. A total of 19 healthy controls (HC) and 16 SMC subjects were included in the present study. Neural responses to the auditory paired-stimulus paradigm were recorded by the magnetoencephalography and analyzed by the distributed source imaging method of minimum norm estimate. The SG of M50 and M100 components were measured using the amplitude ratio of the second response over the first response at the cortical level. Compared to HC, subjects with SMC showed significantly increased M50 SG ratios in the inferior parietal lobule (IPL). Furthermore, M50 SG ratios in the right IPL yielded an acceptable discriminative ability to distinguish SMC from HC. However, we did not find a significant association between SG ratios and cognitive function requiring inhibitory control either in the HC or SMC group. In conclusion, although SMC subjects have intact cognitive functioning revealed by objective neuropsychological tests, their deficits in automatic inhibitory function could be detected through neurophysiological recordings. Our results suggest that altered brain function occurs in SMC prior to the obvious decline of cognitive performance.
Collapse
Affiliation(s)
- Hua-Hsuan Sun
- Department of Occupational Therapy and Graduate Institute of Behavioral Sciences, Chang Gung University, Taoyuan, Taiwan.,Laboratory of Brain Imaging and Neural Dynamics (BIND Lab), Chang Gung University, Taoyuan, Taiwan.,Bali Psychiatric Center, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Mei-Yin Lin
- Department of Physical Medicine and Rehabilitation, Taichung Hospital, Ministry of Health and Welfare, Taichung, Taiwan
| | - Rui Nouchi
- Department of Cognitive Health Science, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Japan.,Smart Aging Research Center (S.A.R.C), Tohoku University, Sendai, Japan
| | - Pei-Ning Wang
- Division of General Neurology, Department of Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Neurology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Hsiung Cheng
- Department of Occupational Therapy and Graduate Institute of Behavioral Sciences, Chang Gung University, Taoyuan, Taiwan.,Laboratory of Brain Imaging and Neural Dynamics (BIND Lab), Chang Gung University, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.,Department of Psychiatry, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
34
|
Buckley RF. Recent Advances in Imaging of Preclinical, Sporadic, and Autosomal Dominant Alzheimer's Disease. Neurotherapeutics 2021; 18:709-727. [PMID: 33782864 PMCID: PMC8423933 DOI: 10.1007/s13311-021-01026-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Observing Alzheimer's disease (AD) pathological changes in vivo with neuroimaging provides invaluable opportunities to understand and predict the course of disease. Neuroimaging AD biomarkers also allow for real-time tracking of disease-modifying treatment in clinical trials. With recent neuroimaging advances, along with the burgeoning availability of longitudinal neuroimaging data and big-data harmonization approaches, a more comprehensive evaluation of the disease has shed light on the topographical staging and temporal sequencing of the disease. Multimodal imaging approaches have also promoted the development of data-driven models of AD-associated pathological propagation of tau proteinopathies. Studies of autosomal dominant, early sporadic, and late sporadic courses of the disease have shed unique insights into the AD pathological cascade, particularly with regard to genetic vulnerabilities and the identification of potential drug targets. Further, neuroimaging markers of b-amyloid, tau, and neurodegeneration have provided a powerful tool for validation of novel fluid cerebrospinal and plasma markers. This review highlights some of the latest advances in the field of human neuroimaging in AD across these topics, particularly with respect to positron emission tomography and structural and functional magnetic resonance imaging.
Collapse
Affiliation(s)
- Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital & Brigham and Women's, Harvard Medical School, Boston, MA, USA.
- Melbourne School of Psychological Sciences and Florey Institutes, University of Melbourne, Melbourne, VIC, Australia.
- Department of Neurology, Massachusetts General Hospital, 149 13th St, Charlestown, MA, 02129, USA.
| |
Collapse
|
35
|
2020 update on the clinical validity of cerebrospinal fluid amyloid, tau, and phospho-tau as biomarkers for Alzheimer's disease in the context of a structured 5-phase development framework. Eur J Nucl Med Mol Imaging 2021; 48:2121-2139. [PMID: 33674895 PMCID: PMC8175301 DOI: 10.1007/s00259-021-05258-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022]
Abstract
Purpose In the last decade, the research community has focused on defining reliable biomarkers for the early detection of Alzheimer’s disease (AD) pathology. In 2017, the Geneva AD Biomarker Roadmap Initiative adapted a framework for the systematic validation of oncological biomarkers to cerebrospinal fluid (CSF) AD biomarkers—encompassing the 42 amino-acid isoform of amyloid-β (Aβ42), phosphorylated-tau (P-tau), and Total-tau (T-tau)—with the aim to accelerate their development and clinical implementation. The aim of this work is to update the current validation status of CSF AD biomarkers based on the Biomarker Roadmap methodology. Methods A panel of experts in AD biomarkers convened in November 2019 at a 2-day workshop in Geneva. The level of maturity (fully achieved, partly achieved, preliminary evidence, not achieved, unsuccessful) of CSF AD biomarkers was assessed based on the Biomarker Roadmap methodology before the meeting and presented and discussed during the workshop. Results By comparison to the previous 2017 Geneva Roadmap meeting, the primary advances in CSF AD biomarkers have been in the area of a unified protocol for CSF sampling, handling and storage, the introduction of certified reference methods and materials for Aβ42, and the introduction of fully automated assays. Additional advances have occurred in the form of defining thresholds for biomarker positivity and assessing the impact of covariates on their discriminatory ability. Conclusions Though much has been achieved for phases one through three, much work remains in phases four (real world performance) and five (assessment of impact/cost). To a large degree, this will depend on the availability of disease-modifying treatments for AD, given these will make accurate and generally available diagnostic tools key to initiate therapy. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05258-7.
Collapse
|
36
|
Pascoal TA, Therriault J, Benedet AL, Savard M, Lussier FZ, Chamoun M, Tissot C, Qureshi MNI, Kang MS, Mathotaarachchi S, Stevenson J, Hopewell R, Massarweh G, Soucy JP, Gauthier S, Rosa-Neto P. 18F-MK-6240 PET for early and late detection of neurofibrillary tangles. Brain 2021; 143:2818-2830. [PMID: 32671408 DOI: 10.1093/brain/awaa180] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/30/2020] [Accepted: 04/14/2020] [Indexed: 11/15/2022] Open
Abstract
Braak stages of tau neurofibrillary tangle accumulation have been incorporated in the criteria for the neuropathological diagnosis of Alzheimer's disease. It is expected that Braak staging using brain imaging can stratify living individuals according to their individual patterns of tau deposition, which may prove crucial for clinical trials and practice. However, previous studies using the first-generation tau PET agents have shown a low sensitivity to detect tau pathology in areas corresponding to early Braak histopathological stages (∼20% of cognitively unimpaired elderly with tau deposition in regions corresponding to Braak I-II), in contrast to ∼80-90% reported in post-mortem cohorts. Here, we tested whether the novel high affinity tau tangles tracer 18F-MK-6240 can better identify individuals in the early stages of tau accumulation. To this end, we studied 301 individuals (30 cognitively unimpaired young, 138 cognitively unimpaired elderly, 67 with mild cognitive impairment, 54 with Alzheimer's disease dementia, and 12 with frontotemporal dementia) with amyloid-β 18F-NAV4694, tau 18F-MK-6240, MRI, and clinical assessments. 18F-MK-6240 standardized uptake value ratio images were acquired at 90-110 min after the tracer injection. 18F-MK-6240 discriminated Alzheimer's disease dementia from mild cognitive impairment and frontotemporal dementia with high accuracy (∼85-100%). 18F-MK-6240 recapitulated topographical patterns consistent with the six hierarchical stages proposed by Braak in 98% of our population. Cognition and amyloid-β status explained most of the Braak stages variance (P < 0.0001, R2 = 0.75). No single region of interest standardized uptake value ratio accurately segregated individuals into the six topographic Braak stages. Sixty-eight per cent of the cognitively unimpaired elderly amyloid-β-positive and 37% of the cognitively unimpaired elderly amyloid-β-negative subjects displayed tau deposition, at least in the transentorhinal cortex (Braak I). Tau deposition solely in the transentorhinal cortex was associated with an elevated prevalence of amyloid-β, neurodegeneration, and cognitive impairment (P < 0.0001). 18F-MK-6240 deposition in regions corresponding to Braak IV-VI was associated with the highest prevalence of neurodegeneration, whereas in Braak V-VI regions with the highest prevalence of cognitive impairment. Our results suggest that the hierarchical six-stage Braak model using 18F-MK-6240 imaging provides an index of early and late tau accumulation as well as disease stage in preclinical and symptomatic individuals. Tau PET Braak staging using high affinity tracers has the potential to be incorporated in the diagnosis of living patients with Alzheimer's disease in the near future.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Muhammad Naveed Iqbal Qureshi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Robert Hopewell
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Gassan Massarweh
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
37
|
Wang Z, Yang J, Zhu W, Tang Y, Jia J. The synaptic marker neurogranin as a disease state biomarker in Alzheimer's disease: a systematic review and meta-analysis. Int J Neurosci 2021; 132:1245-1253. [PMID: 33527855 DOI: 10.1080/00207454.2021.1881087] [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: 10/22/2022]
Abstract
Objective: Synaptic degeneration is the pathologic foundation of cognitive decline in the Alzheimer's disease (AD) continuum. We aimed to determine whether cerebrospinal fluid (CSF) synaptic marker neurogranin (Ng) is a disease state or a disease stage biomarker in the AD continuum.Methods: Studies comparing CSF Ng levels among AD, mild cognitive impairment (MCI) and healthy participants were included. Studies were eligible if the correlation between CSF Ng levels and Mini-Mental Status Examination (MMSE) scores was investigated.Results: Twenty-one studies met our inclusion criteria (n = 4515). The magnitude of effect sizes was more apparent in AD (standardized mean difference [SMD] = 1.72; 95% confidence interval [CI] = 1.23-2.22), than in MCI (SMD = 0.82; 95% CI = 0.29-1.34) compared to control populations. These results suggest that CSF Ng can discriminate AD and MCI from control populations, implying that synaptic degeneration worsens as patients progress from MCI to AD. However, there was a very weak correlation between CSF Ng levels and MMSE scores (r = -0.15; 95% CI = -0.21--0.08) among the whole populations, suggesting that an increment of CSF Ng is best considered a biological evidence of disease state in the AD continuum.Conclusion: Our study provides evidence that the synaptic marker CSF Ng can be used as a disease state biomarker for the AD continuum. Because synaptic degeneration is a distinct pathologic event from amyloid deposition and neurofibrillary tangle formation, CSF Ng may provide an important supplementation to the AT(N) biomarker system to reveal the sequence of neuropathology.
Collapse
Affiliation(s)
- Zhibin Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jianwei Yang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Wei Zhu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China.,Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
38
|
Day GS, Gordon BA, McCullough A, Bucelli RC, Perrin RJ, Benzinger TLS, Ances BM. Flortaucipir (tau) PET in LGI1 antibody encephalitis. Ann Clin Transl Neurol 2021; 8:491-497. [PMID: 33410601 PMCID: PMC7886030 DOI: 10.1002/acn3.51297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 01/05/2023] Open
Abstract
The contributors to persistent cognitive impairment and hippocampal atrophy in leucine-rich glioma-inactivated 1 antibody encephalitis (LGI1) patients are unknown. We evaluated whether tau neuropathology measured with [18 F]flortaucipir PET neuroimaging associated with persistent cognitive impairment and hippocampal atrophy in four recovering LGI1 patients (3 men; median age, 67 [37-88] years). Imaging findings in cases were compared with those observed in age- and gender-similar cognitively normal individuals (n = 124) and individuals with early-symptomatic Alzheimer disease (n = 11). Elevated [18 F]flortaucipir retention was observed in the two LGI1 patients with hippocampal atrophy and persistent cognitive impairment, including one with autopsy-confirmed Alzheimer disease. Tau neuropathology may associate with cognitive complaints and hippocampal atrophy in recovering LGI1 patients.
Collapse
Affiliation(s)
- Gregory S. Day
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
| | - Brian A. Gordon
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
| | - Austin McCullough
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
| | - Robert C. Bucelli
- Washington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine in Saint Louis JacksonvilleSaint LouisMissouriUSA
| | - Richard J. Perrin
- Washington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine in Saint Louis JacksonvilleSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University in Saint Louis School of MedicineSaint LouisMissouriUSA
| | - Tammie L. S. Benzinger
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
| | - Beau M. Ances
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine in Saint Louis JacksonvilleSaint LouisMissouriUSA
| |
Collapse
|
39
|
Meeker KL, Wisch JK, Hudson D, Coble D, Xiong C, Babulal GM, Gordon BA, Schindler SE, Cruchaga C, Flores S, Dincer A, Benzinger TL, Morris JC, Ances BM. Socioeconomic Status Mediates Racial Differences Seen Using the AT(N) Framework. Ann Neurol 2021; 89:254-265. [PMID: 33111990 PMCID: PMC7903892 DOI: 10.1002/ana.25948] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVES African Americans are at greater risk for developing Alzheimer's disease (AD) dementia than non-Hispanic whites. In addition to biological considerations (eg, genetic influences and comorbid disorders), social and environmental factors may increase the risk of AD dementia. This paper (1) assesses neuroimaging biomarkers of amyloid (A), tau (T), and neurodegeneration (N) for potential racial differences and (2) considers mediating effects of socioeconomic status (SES) and measures of small vessel and cardiovascular disease on observed race differences. METHODS Imaging measures of AT(N) (amyloid and tau positron emission tomography [PET]) structural magnetic resonance imaging (MRI), and resting state functional connectivity (rs-fc) were collected from African American (n = 131) and white (n = 685) cognitively normal participants age 45 years and older. Measures of small vessel and cardiovascular disease (white matter hyperintensities [WMHs] on MRI, blood pressure, and body mass index [BMI]) and area-based SES were included in mediation analyses. RESULTS Compared to white participants, African American participants had greater neurodegeneration, as measured by decreased cortical volumes (Cohen's f2 = 0.05, p < 0.001). SES mediated the relationship between race and cortical volumes. There were no significant race effects for amyloid, tau, or rs-fc signature. INTERPRETATION Modifiable factors, such as differences in social contexts and resources, particularly area-level SES, may contribute to observed racial differences in AD. Future studies should emphasize collection of relevant psychosocial factors in addition to the development of intentional diversity and inclusion efforts to improve the racial/ethnic and socioeconomic representativeness of AD studies. ANN NEUROL 2021;89:254-265.
Collapse
Affiliation(s)
- Karin L Meeker
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie K Wisch
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Darrell Hudson
- Brown School, Washington University in St. Louis, St. Louis, MO, USA
| | - Dean Coble
- Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ganesh M Babulal
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Shaney Flores
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Aylin Dincer
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Tammie L Benzinger
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
40
|
Ayodele T, Rogaeva E, Kurup JT, Beecham G, Reitz C. Early-Onset Alzheimer's Disease: What Is Missing in Research? Curr Neurol Neurosci Rep 2021; 21:4. [PMID: 33464407 PMCID: PMC7815616 DOI: 10.1007/s11910-020-01090-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Early-onset Alzheimer's disease (EOAD), defined as Alzheimer's disease (AD) occurring before age 65, is significantly less well studied than the late-onset form (LOAD) despite EOAD often presenting with a more aggressive disease progression. The aim of this review is to summarize the current understanding of the etiology of EOAD, their translation into clinical practice, and to suggest steps to be taken to move our understanding forward. RECENT FINDINGS EOAD cases make up 5-10% of AD cases but only 10-15% of these cases show known mutations in the APP, PSEN1, and PSEN2, which are linked to EOAD. New data suggests that these unexplained cases following a non-Mendelian pattern of inheritance is potentially caused by a mix of common and newly discovered rare variants. However, only a fraction of this genetic variation has been identified to date leaving the molecular mechanisms underlying this type of AD and their association with clinical, biomarker, and neuropathological changes unclear. While great advancements have been made in characterizing EOAD, much work is needed to disentangle the molecular mechanisms underlying this type of AD and to identify putative targets for more precise disease screening, diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Temitope Ayodele
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada
| | - Jiji T Kurup
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Gary Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Christiane Reitz
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA.
- Department of Neurology, Columbia University, New York, NY, USA.
- Department of Epidemiology, Sergievsky Center, Taub Institute for Research on the Aging Brain, Columbia University, 630 W 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
41
|
Radioactive synthesis of tau PET imaging agent 18F-AV-1451 and its role in monitoring the progression of Alzheimer's disease and supporting differential diagnosis. Ann Nucl Med 2021; 35:139-147. [PMID: 33460010 DOI: 10.1007/s12149-020-01566-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
Alzheimer's disease (AD) is on the rise all over the world, and brings with it great challenges to medical care and heavy burdens to family and society. Accurate diagnosis and differential diagnosis are of great importance. Tau positron emission tomography (PET) might offer novel insights and be of great assistance in monitoring disease progression and supporting the differential diagnosis. 18F-AV-1451, as the first Tau PET imaging agent approved by the Food and Drug Administration (FDA), has been of great potential in clinical trials. Here, we reviewed the synthesis and characteristics of 18F-AV-1451 and its role in monitoring AD progression and supporting the differential diagnosis.
Collapse
|
42
|
Provost K, Iaccarino L, Soleimani-Meigooni DN, Baker S, Edwards L, Eichenlaub U, Hansson O, Jagust W, Janabi M, La Joie R, Lesman-Segev O, Mellinger TJ, Miller BL, Ossenkoppele R, Pham J, Smith R, Sonni I, Strom A, Mattsson-Carlgren N, Rabinovici GD. Comparing ATN-T designation by tau PET visual reads, tau PET quantification, and CSF PTau181 across three cohorts. Eur J Nucl Med Mol Imaging 2021; 48:2259-2271. [PMID: 33398408 DOI: 10.1007/s00259-020-05152-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/06/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE To compare rates of tau biomarker positivity (T-status) per the 2018 Alzheimer's Disease (AD) Research Framework derived from [18F]flortaucipir (FTP) PET visual assessment, FTP quantification, and cerebrospinal fluid (CSF) phosphorylated Tau-181 (PTau181). METHODS We included 351 subjects with varying clinical diagnoses from three cohorts with available FTP PET and CSF PTau181 within 18 months. T-status was derived from (1) FTP visual assessment by two blinded raters; (2) FTP standardized uptake value ratio (SUVR) quantification from a temporal meta-ROI (threshold: SUVR ≥1.27); and (3) Elecsys® Phospho-Tau (181P) CSF (Roche Diagnostics) concentrations (threshold: PTau181 ≥ 24.5 pg/mL). RESULTS FTP visual reads yielded the highest rates of T+, while T+ by SUVR increased progressively from cognitively normal (CN) through mild cognitive impairment (MCI) and AD dementia. T+ designation by CSF PTau181 was intermediate between FTP visual reads and SUVR values in CN, similar to SUVR in MCI, and lower in AD dementia. Concordance in T-status between modality pairs ranged from 68 to 76% and varied by clinical diagnosis, being highest in patients with AD dementia. In discriminating Aβ + MCI and AD subjects from healthy controls and non-AD participants, FTP visual assessment was most sensitive (0.96) but least specific (0.60). Specificity was highest with FTP SUVR (0.91) with sensitivity of 0.89. Sensitivity (0.73) and specificity (0.72) were balanced for PTau181. CONCLUSION The choice of tau biomarker may differ by disease stage and research goals that seek to maximize sensitivity or specificity. Visual interpretations of tau PET enhance sensitivity compared to quantification alone, particularly in early disease stages.
Collapse
Affiliation(s)
- Karine Provost
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA.
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Suzanne Baker
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | | | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - William Jagust
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA, USA
| | - Mustafa Janabi
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Orit Lesman-Segev
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Taylor J Mellinger
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Julie Pham
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Ida Sonni
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, UC Los Angeles, Los Angeles, CA, USA
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
43
|
Duits FH, Wesenhagen KEJ, Ekblad L, Wolters E, Willemse EAJ, Scheltens P, van der Flier WM, Teunissen CE, Visser PJ, Tijms BM. Four subgroups based on tau levels in Alzheimer's disease observed in two independent cohorts. Alzheimers Res Ther 2021; 13:2. [PMID: 33397464 PMCID: PMC7780683 DOI: 10.1186/s13195-020-00713-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND As Alzheimer's disease (AD) pathology presents decades before dementia manifests, unbiased biomarker cut-points may more closely reflect presence of pathology than clinically defined cut-points. Currently, unbiased cerebrospinal fluid (CSF) tau cut-points are lacking. METHODS We investigated CSF t-tau and p-tau cut-points across the clinical spectrum using Gaussian mixture modelling, in two independent cohorts (Amsterdam Dementia Cohort and ADNI). RESULTS Individuals with normal cognition (NC) (total n = 1111), mild cognitive impairment (MCI) (total n = 1213) and Alzheimer's disease dementia (AD) (total n = 1524) were included. In both cohorts, four CSF t- and p-tau distributions and three corresponding cut-points were identified. Increasingly high tau subgroups were characterized by steeper MMSE decline and higher progression risk to AD (cohort/platform-dependent HR, t-tau 1.9-21.3; p-tau 2.2-9.5). LIMITATIONS The number of subjects in some subgroups and subanalyses was small, especially in the highest tau subgroup and in tau PET analyses. CONCLUSIONS In two independent cohorts, t-tau and p-tau levels showed four subgroups. Increasingly high tau subgroups were associated with faster clinical decline, suggesting our approach may aid in more precise prognoses.
Collapse
Affiliation(s)
- Flora H Duits
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Kirsten E J Wesenhagen
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Laura Ekblad
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Emma Wolters
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Eline A J Willemse
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Pieter Jelle Visser
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Alzheimer Center Limburg, Department of Psychiatry & Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Betty M Tijms
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
44
|
Abstract
Pathological accumulated misfolded tau underlies various neurodegenerative diseases and associated clinical syndromes. To diagnose those diseases reliable before death or even at early stages, many different tau-specific radiotracers have been developed in the last decade to be used with positron-emission-tomography. In contrast to amyloid-β imaging, different isoforms of tau exist further complicating radiotracer development. First-generation radiotracers like [11C]PBB3, [18F]AV1451 and [18F]THK5351 have been extensively investigated in vitro and in vivo. In Alzheimer's disease (AD), high specific binding could be detected, and evidence of clinical applicability recently led to clinical approval of [18F]flortaucipir ([18F]AV1451) by the FDA. Nevertheless, absent or minor binding to non-AD tau isoforms and high off-target binding to non-tau brain structures limit the diagnostic applicability especially in non-AD tauopathies demanding further tracer development. In vitro assays and autoradiography results of next-generation radiotracers [18F]MK-6240, [18F]RO-948, [18F]PM-PBB3, [18F]GTP-1 and [18F]PI-2620 clearly indicate less off-target binding and high specific binding to tau neurofibrils. First in human studies have been conducted with promising results for all tracers in AD patients, and also some positive experience in non-AD tauopathies. Overall, larger scaled autoradiography and human studies are needed to further evaluate the most promising candidates and support future clinical approval.
Collapse
Affiliation(s)
- Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
45
|
Schultz SA, Gordon BA, Mishra S, Su Y, Morris JC, Ances BM, Duchek JM, Balota DA, Benzinger TL. Association between personality and tau-PET binding in cognitively normal older adults. Brain Imaging Behav 2020; 14:2122-2131. [PMID: 31486975 PMCID: PMC7056533 DOI: 10.1007/s11682-019-00163-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Personality traits such as Neuroticism and Conscientiousness are associated with Alzheimer disease (AD) pathophysiology in cognitively normal (CN) and impaired individuals, and may represent potential risk or resilience factors, respectively. This study examined the cross-sectional relationship between personality traits and regional tau deposition using positron emission tomography (PET) in cognitively normal older adults. A cohort of CN (Clinical Dementia Rating (CDR) 0, n = 128) older adults completed the NEO Five-Factor Inventory to assess traits of Neuroticism, Extroversion, Openness, Agreeableness, and Conscientiousness and underwent tau-PET and β-amyloid (Aβ)-PET imaging. We utilized linear regression models, adjusting for age, sex, geriatric depression score, and Aβ to evaluate the association between each of the personality traits and regional tau-PET accumulation. Elevated Neuroticism scores were associated with higher tau-PET accumulation in the amygdala (p = .002), entorhinal cortex (p = .012), and inferior temporal cortex (p = .016), as well as with a composite tau-PET measure (p = .002). In contrast, Extroversion, Openness, Agreeableness, and Conscientiousness were not associated with tau deposition in any of these regions (p's > 0.160). Our results indicate that increased Neuroticism is associated with higher tau pathophysiology in regions known to be vulnerable to AD pathophysiology in CN participants. High Neuroticism scores may therefore serve as a potential risk factor for tau accumulation. Alternatively, personality can change with the onset of AD, thus increased tau levels may affect Neuroticism scores. While future longitudinal studies are needed to determine directionality, our findings suggest early associations between Neuroticism and tau accumulation in CN adults.
Collapse
Affiliation(s)
- Stephanie A. Schultz
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO.,Department of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Department of Psychological & Brain Sciences, Washington University, St. Louis, MO
| | - Shruti Mishra
- Department of Radiology, Washington University School of Medicine, St. Louis, MO.,Department of Radiology, Mass General Hospital, Boston, MA
| | - Yi Su
- Department of Radiology, Washington University School of Medicine, St. Louis, MO.,Banner Alzheimer’s Institute, Phoenix, AZ
| | - John C. Morris
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Beau M. Ances
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Janet M. Duchek
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Department of Psychological & Brain Sciences, Washington University, St. Louis, MO
| | - David A. Balota
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Department of Psychological & Brain Sciences, Washington University, St. Louis, MO
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
46
|
Mukherjee S, Heath L, Preuss C, Jayadev S, Garden GA, Greenwood AK, Sieberts SK, De Jager PL, Ertekin-Taner N, Carter GW, Mangravite LM, Logsdon BA. Molecular estimation of neurodegeneration pseudotime in older brains. Nat Commun 2020; 11:5781. [PMID: 33188183 PMCID: PMC7666177 DOI: 10.1038/s41467-020-19622-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/23/2020] [Indexed: 01/15/2023] Open
Abstract
The temporal molecular changes that lead to disease onset and progression in Alzheimer's disease (AD) are still unknown. Here we develop a temporal model for these unobserved molecular changes with a manifold learning method applied to RNA-Seq data collected from human postmortem brain samples collected within the ROS/MAP and Mayo Clinic RNA-Seq studies. We define an ordering across samples based on their similarity in gene expression and use this ordering to estimate the molecular disease stage-or disease pseudotime-for each sample. Disease pseudotime is strongly concordant with the burden of tau (Braak score, P = 1.0 × 10-5), Aβ (CERAD score, P = 1.8 × 10-5), and cognitive diagnosis (P = 3.5 × 10-7) of late-onset (LO) AD. Early stage disease pseudotime samples are enriched for controls and show changes in basic cellular functions. Late stage disease pseudotime samples are enriched for late stage AD cases and show changes in neuroinflammation and amyloid pathologic processes. We also identify a set of late stage pseudotime samples that are controls and show changes in genes enriched for protein trafficking, splicing, regulation of apoptosis, and prevention of amyloid cleavage pathways. In summary, we present a method for ordering patients along a trajectory of LOAD disease progression from brain transcriptomic data.
Collapse
Affiliation(s)
- Sumit Mukherjee
- Sage Bionetworks, Seattle, WA, USA
- Microsoft, Redmond, WA, USA
| | | | | | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Gwenn A Garden
- Department of Neurology, University of Washington, Seattle, WA, USA
| | | | | | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York City, NY, USA
- Taub Institute, Columbia University Irving Medical Center, New York City, NY, USA
| | - Nilüfer Ertekin-Taner
- Department of Neurology, Mayo Clinic Florid, Jacksonville, FL, USA
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | | | | | - Benjamin A Logsdon
- Sage Bionetworks, Seattle, WA, USA.
- Cajal Neuroscience, Seattle, WA, USA.
| |
Collapse
|
47
|
Suárez-Calvet M, Karikari TK, Ashton NJ, Lantero Rodríguez J, Milà-Alomà M, Gispert JD, Salvadó G, Minguillon C, Fauria K, Shekari M, Grau-Rivera O, Arenaza-Urquijo EM, Sala-Vila A, Sánchez-Benavides G, González-de-Echávarri JM, Kollmorgen G, Stoops E, Vanmechelen E, Zetterberg H, Blennow K, Molinuevo JL. Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer's continuum when only subtle changes in Aβ pathology are detected. EMBO Mol Med 2020; 12:e12921. [PMID: 33169916 PMCID: PMC7721364 DOI: 10.15252/emmm.202012921] [Citation(s) in RCA: 222] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023] Open
Abstract
In Alzheimer's disease (AD), tau phosphorylation in the brain and its subsequent release into cerebrospinal fluid (CSF) and blood is a dynamic process that changes during disease evolution. The main aim of our study was to characterize the pattern of changes in phosphorylated tau (p-tau) in the preclinical stage of the Alzheimer's continuum. We measured three novel CSF p-tau biomarkers, phosphorylated at threonine-181 and threonine-217 with an N-terminal partner antibody and at threonine-231 with a mid-region partner antibody. These were compared with an automated mid-region p-tau181 assay (Elecsys) as the gold standard p-tau measure. We demonstrate that these novel p-tau biomarkers increase more prominently in preclinical Alzheimer, when only subtle changes of amyloid-β (Aβ) pathology are detected, and can accurately differentiate Aβ-positive from Aβ-negative cognitively unimpaired individuals. Moreover, we show that the novel plasma N-terminal p-tau181 biomarker is mildly but significantly increased in the preclinical stage. Our results support the idea that early changes in neuronal tau metabolism in preclinical Alzheimer, likely in response to Aβ exposure, can be detected with these novel p-tau assays.
Collapse
Affiliation(s)
- Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Wallenberg Centre for Molecular and Translational Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, Psychology & Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Juan Lantero Rodríguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain.,Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Eider M Arenaza-Urquijo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Aleix Sala-Vila
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - José Maria González-de-Echávarri
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | | | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 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
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de sFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | | |
Collapse
|
48
|
Rostamzadeh A, Jessen F. [Early detection of Alzheimer's disease and dementia prediction in patients with mild cognitive impairment : Summary of current recommendations]. DER NERVENARZT 2020; 91:832-842. [PMID: 32300816 DOI: 10.1007/s00115-020-00907-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mild cognitive impairment (MCI) is characterized by cognitive deficits but essentially preserved competence in activities of daily living. It is a risk factor for the development of dementia and can reflect a prodromal predementia state of Alzheimer's disease (AD). The pathology of AD is defined by cerebral deposition of amyloid-beta-1-42 protein and aggregation of phosphorylated tau protein, which can be identified in vivo by biomarkers for these alterations. As a result of advances in the field of biomarker-based early detection of AD, it is possible to differentiate between MCI patients with and without a pathological AD condition and therefore, between patients with a low and those with a high risk for the development of dementia. At present there are no specific guideline recommendations in Germany for the diagnostic use of biomarkers in predementia detection of AD and for dementia risk assessment in patients with MCI. This article summarizes the current recommendations of a European expert consensus publication and a multidisciplinary working group of the Alzheimer's Association on the clinical application of cerebrospinal fluid (CSF) biomarkers for the diagnostics of AD in patients with MCI. If the clinical diagnostic criteria for MCI are fulfilled according to the medical history and neuropsychological testing, it is recommended to carry out further diagnostics (blood test, brain imaging) in order to more precisely define the differential diagnostic classification. Counseling on the potential benefits, limits and risks of biomarker testing for early AD detection and dementia risk prediction should always precede assessment of CSF biomarkers. Information about the individual risk of developing dementia has potential consequences for the psychological well-being and life planning; therefore, clinical follow-up visits are recommended.
Collapse
Affiliation(s)
- Ayda Rostamzadeh
- Klinik für Psychiatrie und Psychotherapie, Uniklinik Köln, Medizinische Fakultät, Köln, Deutschland.
| | - Frank Jessen
- Klinik für Psychiatrie und Psychotherapie, Uniklinik Köln, Medizinische Fakultät, Köln, Deutschland.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Deutschland
| |
Collapse
|
49
|
Guo Y, Li H, Tan L, Chen S, Yang Y, Ma Y, Zuo C, Dong Q, Tan L, Yu J. Discordant Alzheimer's neurodegenerative biomarkers and their clinical outcomes. Ann Clin Transl Neurol 2020; 7:1996-2009. [PMID: 32949193 PMCID: PMC7545611 DOI: 10.1002/acn3.51196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/08/2020] [Accepted: 08/25/2020] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE In the 2018 ATN framework, Alzheimer's neurodegenerative biomarkers comprised cerebrospinal fluid (CSF) total tau, 18 F-fluorodeoxyglucose-positron emission tomography, and brain atrophy. We aimed to assess the clinical outcomes of having discordant Alzheimer's neurodegenerative biomarkers. METHODS A total of 721 non-demented individuals from the Alzheimer's Disease Neuroimaging Initiative database were included and then further categorized into concordant-negative, discordant, and concordant-positive groups. Demographic distributions of the groups were compared. Longitudinal changes in clinical outcomes and risk of conversion were assessed using linear mixed-effects models and multivariate Cox proportional hazard models, respectively. RESULTS Discordant group was intermediate to concordant-negative and concordant-positive groups in terms of APOE ε4 positivity, CSF amyloid-beta, and phosphorylated tau. Compared with concordant-negative group, discordant group deteriorated faster in cognitive scores (Mini-Mental State Examination, the Clinical Dementia Rating Scale-Sum of Boxes, and the Functional Activities Questionnaire) and demonstrated greater rates of atrophy in brain structures (hippocampus, entorhinal cortex, and whole brain), and concordant-positive group performed worse over time than discordant group. Moreover, the risk of cognitive decline increased from concordant-negative to discordant to concordant-positive. The results from longitudinal analyses were validated in A+T+, cognitively normal, and mild cognitive impairment individuals, and were also validated by applying different cutoffs and neurodegenerative biomarkers. INTERPRETATION Discordant neurodegenerative status denotes a stage of cognitive function which is intermediate between concordant-negative and concordant-positive. Identification of discordant cases would provide insights into intervention and new therapy approaches, particularly in A+T+ individuals. Moreover, this work may be a complement to the ATN scheme.
Collapse
Affiliation(s)
- Yu Guo
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Hong‐Qi Li
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Lin Tan
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Shi‐Dong Chen
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Yu‐Xiang Yang
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Ya‐Hui Ma
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Chuan‐Tao Zuo
- PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Qiang Dong
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Lan Tan
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Jin‐Tai Yu
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | | |
Collapse
|
50
|
Kim HR, Lee T, Choi JK, Jeong Y. Genetic variants beyond amyloid and tau associated with cognitive decline: A cohort study. Neurology 2020; 95:e2366-e2377. [PMID: 32938779 DOI: 10.1212/wnl.0000000000010724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 04/27/2020] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE To identify single nucleotide polymorphisms (SNPs) associated with cognitive decline independent of β-amyloid (Aβ) and tau pathology in Alzheimer disease (AD). METHODS Discovery and replication datasets consisting of 414 individuals (94 cognitively normal control [CN], 185 with mild cognitive impairment [MCI], and 135 with AD) and 72 individuals (22 CN, 39 with MCI, and 11 with AD), respectively, were obtained from the Alzheimer's Disease Neuroimaging Initiative database. Genome-wide association analysis was conducted to identify SNPs associated with individual cognitive function (measured with the Mini-Mental State Examination and Alzheimer's Disease Assessment Scale-Cognitive Subscale ) while controlling for the level of Aβ and tau (measured as CSF phosphorylated-tau/Aβ1-42). Gene ontology analysis was performed on SNP-associated genes. RESULTS We identified 1 significant (rs55906536, β = -1.91, standard error 0.34, p = 4.07 × 10-8) and 4 suggestive variants on chromosome 6 that were associated with poorer cognitive function. Congruent results were found in the replication data. A structural equation model showed that the identified SNP deteriorated cognitive function partially through cortical thinning of the brain in a region-specific manner. Furthermore, a bioinformatics analysis showed that the identified SNPs were associated with genes related to glutathione metabolism. CONCLUSIONS In this study, we identified SNPs related to cognitive decline in a manner that could not be explained by Aβ and tau levels. Our findings provide insight into the complexity of AD pathogenesis and support the growing literature on the role of glutathione in AD.
Collapse
Affiliation(s)
- Hang-Rai Kim
- From the Graduate School of Medical Science & Engineering (H.-R.K., T.L., Y.J.), KAIST Institute for Health Science and Technology (H.-R.K., Y.J.), and Department of Bio and Brain Engineering (J.K.C., Y.J.), KAIST, Daejeon, Republic of Korea
| | - Taeyeop Lee
- From the Graduate School of Medical Science & Engineering (H.-R.K., T.L., Y.J.), KAIST Institute for Health Science and Technology (H.-R.K., Y.J.), and Department of Bio and Brain Engineering (J.K.C., Y.J.), KAIST, Daejeon, Republic of Korea
| | - Jung Kyoon Choi
- From the Graduate School of Medical Science & Engineering (H.-R.K., T.L., Y.J.), KAIST Institute for Health Science and Technology (H.-R.K., Y.J.), and Department of Bio and Brain Engineering (J.K.C., Y.J.), KAIST, Daejeon, Republic of Korea
| | - Yong Jeong
- From the Graduate School of Medical Science & Engineering (H.-R.K., T.L., Y.J.), KAIST Institute for Health Science and Technology (H.-R.K., Y.J.), and Department of Bio and Brain Engineering (J.K.C., Y.J.), KAIST, Daejeon, Republic of Korea.
| | | |
Collapse
|