101
|
García-Escobar G, Puig-Pijoan A, Puente-Periz V, Fernández-Lebrero A, María Manero R, Navalpotro-Gómez I, Suárez-Calvet M, Grau-Rivera O, Contador-Muñana J, Cascales-Lahoz D, Duran-Jordà X, Boltes N, Pont-Sunyer MC, Ortiz-Gil J, Carrillo-Molina S, López-Villegas MD, Abellán-Vidal MT, Martínez-Casamitjana MI, Hernández-Sánchez JJ, Padrós-Fluvià A, Peña-Casanova J, Sánchez-Benavides G. NEURONORMA Cognitive Battery Associations with Cerebrospinal Fluid Amyloid-β and Tau Levels in the Continuum of Alzheimer's Disease. J Alzheimers Dis 2023; 92:1303-1321. [PMID: 37038810 DOI: 10.3233/jad-220930] [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: 04/12/2023]
Abstract
BACKGROUND Neuropsychological assessments are essential to define the cognitive profile and contribute to the diagnosis of Alzheimer's disease (AD). The progress in knowledge about the pathophysiological process of the disease has allowed conceptualizing AD through biomarkers as a biological continuum that encompasses different clinical stages. OBJECTIVE To explore the association between cerebrospinal fluid (CSF) biomarkers of AD and cognition using the NEURONORMA battery, in a sample of cognitively unimpaired (CU), mild cognitive impaired (MCI), and mild dementia of the Alzheimer type (DAT) subjects, and to characterize the cognitive profiles in MCI subjects classified by A/T/N system. METHODS 42 CU, 35 MCI, and 35 mild DAT were assessed using the NEURONORMA battery. Core AD biomarkers [amyloid-β42 (Aβ42) peptide, total tau (t-tau), and phosphorylated tau 181 (p-tau181)] proteins were measured in CSF. Correlation coefficients, multivariate regression, and effect sizes were calculated. We explored the age- and education-adjusted cognitive profiles by A/T/N variants within the MCI group. RESULTS Cognitive outcomes were directly associated with CSF Aβ42 and inversely with CSF tau measures. We found differences in both biomarkers and cognitive outcomes comparing all pairs except for CSF measures between cognitively impaired groups. The highest effect size was in memory tasks and biomarkers ratios. Lower performances were in memory and executive domains in MCI subjects with AD pathology (A+T+N±) compared to those with normal levels of AD biomarkers (A- T- N). CONCLUSION This study provides further evidence of the validity of Spanish NEURONORMA cognitive battery to characterize cognitive impairment in the AD pathological continuum.
Collapse
Affiliation(s)
- Greta García-Escobar
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Albert Puig-Pijoan
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Víctor Puente-Periz
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Aida Fernández-Lebrero
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Rosa María Manero
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Irene Navalpotro-Gómez
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Marc Suárez-Calvet
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
- 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 Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Oriol Grau-Rivera
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
- 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 Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - José Contador-Muñana
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Diego Cascales-Lahoz
- Cognitive Impairment and Movement Disorders Unit, Neurology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | | | - Núncia Boltes
- Neurology Department, Hospital General de Granollers, Granollers, Spain
| | | | - Jordi Ortiz-Gil
- Neurology Department, Hospital General de Granollers, Granollers, Spain
- Psychology Unit, Hospital General de Granollers, Granollers, Spain
- Maria Angustias Gimenez Research Foundation (FIDMAG), Sant Boi del Llobregat, Spain
| | - Sara Carrillo-Molina
- Neurology Department, Hospital General de Granollers, Granollers, Spain
- Psychology Unit, Hospital General de Granollers, Granollers, Spain
| | - María Dolores López-Villegas
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centre Emili Mira, Institut de Neuropsiquiatria i Addiccions (INAD), Parc de Salut Mar, Santa Coloma de Gramenet, Spain
| | - María Teresa Abellán-Vidal
- Centre Emili Mira, Institut de Neuropsiquiatria i Addiccions (INAD), Parc de Salut Mar, Santa Coloma de Gramenet, Spain
| | | | | | | | - Jordi Peña-Casanova
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), 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 Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| |
Collapse
|
102
|
Long JM, Coble DW, Xiong C, Schindler SE, Perrin RJ, Gordon BA, Benzinger TLS, Grant E, Fagan AM, Harari O, Cruchaga C, Holtzman DM, Morris JC. Preclinical Alzheimer's disease biomarkers accurately predict cognitive and neuropathological outcomes. Brain 2022; 145:4506-4518. [PMID: 35867858 PMCID: PMC10200309 DOI: 10.1093/brain/awac250] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease biomarkers are widely accepted as surrogate markers of underlying neuropathological changes. However, few studies have evaluated whether preclinical Alzheimer's disease biomarkers predict Alzheimer's neuropathology at autopsy. We sought to determine whether amyloid PET imaging or CSF biomarkers accurately predict cognitive outcomes and Alzheimer's disease neuropathological findings. This study included 720 participants, 42-91 years of age, who were enrolled in longitudinal studies of memory and aging in the Washington University Knight Alzheimer Disease Research Center and were cognitively normal at baseline, underwent amyloid PET imaging and/or CSF collection within 1 year of baseline clinical assessment, and had subsequent clinical follow-up. Cognitive status was assessed longitudinally by Clinical Dementia Rating®. Biomarker status was assessed using predefined cut-offs for amyloid PET imaging or CSF p-tau181/amyloid-β42. Subsequently, 57 participants died and underwent neuropathologic examination. Alzheimer's disease neuropathological changes were assessed using standard criteria. We assessed the predictive value of Alzheimer's disease biomarker status on progression to cognitive impairment and for presence of Alzheimer's disease neuropathological changes. Among cognitively normal participants with positive biomarkers, 34.4% developed cognitive impairment (Clinical Dementia Rating > 0) as compared to 8.4% of those with negative biomarkers. Cox proportional hazards modelling indicated that preclinical Alzheimer's disease biomarker status, APOE ɛ4 carrier status, polygenic risk score and centred age influenced risk of developing cognitive impairment. Among autopsied participants, 90.9% of biomarker-positive participants and 8.6% of biomarker-negative participants had Alzheimer's disease neuropathological changes. Sensitivity was 87.0%, specificity 94.1%, positive predictive value 90.9% and negative predictive value 91.4% for detection of Alzheimer's disease neuropathological changes by preclinical biomarkers. Single CSF and amyloid PET baseline biomarkers were also predictive of Alzheimer's disease neuropathological changes, as well as Thal phase and Braak stage of pathology at autopsy. Biomarker-negative participants who developed cognitive impairment were more likely to exhibit non-Alzheimer's disease pathology at autopsy. The detection of preclinical Alzheimer's disease biomarkers is strongly predictive of future cognitive impairment and accurately predicts presence of Alzheimer's disease neuropathology at autopsy.
Collapse
Affiliation(s)
- Justin M Long
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Dean W Coble
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Richard J Perrin
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Brian A Gordon
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Elizabeth Grant
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Anne M Fagan
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Oscar Harari
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - David M Holtzman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
103
|
Tomassen J, den Braber A, van der Lee SJ, Reus LM, Konijnenberg E, Carter SF, Yaqub M, van Berckel BNM, Collij LE, Boomsma DI, de Geus EJC, Scheltens P, Herholz K, Tijms BM, Visser PJ. Amyloid-β and APOE genotype predict memory decline in cognitively unimpaired older individuals independently of Alzheimer's disease polygenic risk score. BMC Neurol 2022; 22:484. [PMID: 36522743 PMCID: PMC9753236 DOI: 10.1186/s12883-022-02925-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND What combination of risk factors for Alzheimer's disease (AD) are most predictive of cognitive decline in cognitively unimpaired individuals remains largely unclear. We studied associations between APOE genotype, AD-Polygenic Risk Scores (AD-PRS), amyloid-β pathology and decline in cognitive functioning over time in a large sample of cognitively unimpaired older individuals. METHODS We included 276 cognitively unimpaired older individuals (75 ± 10 years, 63% female) from the EMIF-AD PreclinAD cohort. An AD-PRS was calculated including 83 genome-wide significant variants. The APOE gene was not included in the PRS and was analyzed separately. Baseline amyloid-β status was assessed by visual read of [18F]flutemetamol-PET standardized uptake value images. At baseline and follow-up (2.0 ± 0.4 years), the cognitive domains of memory, attention, executive function, and language were measured. We used generalized estimating equations corrected for age, sex and center to examine associations between APOE genotype and AD-PRS with amyloid-β status. Linear mixed models corrected for age, sex, center and education were used to examine associations between APOE genotype, AD-PRS and amyloid-β status, and their interaction on changes in cognitive functioning over time. RESULTS Fifty-two participants (19%) had abnormal amyloid-β, and 84 participants (31%) carried at least one APOE ε4 allele. APOE genotype and AD-PRS were both associated with abnormal amyloid-β status. Increasingly more risk-full APOE genotype, a high AD-PRS and an abnormal amyloid-β status were associated with steeper decline in memory functioning in separate models (all p ≤ 0.02). A model including 4-way interaction term (APOE×AD-PRS×amyloid-β×time) was not significant. When modelled together, both APOE genotype and AD-PRS predicted steeper decline in memory functioning (APOE β(SE)=-0.05(0.02); AD-PRS β(SE)=-0.04(0.01)). Additionally, when modelled together, both amyloid-β status and AD-PRS predicted a steeper decline in memory functioning (amyloid-β β(SE)=-0.07(0.04); AD-PRS β(SE)=-0.04(0.01)). Modelling both APOE genotype and amyloid-β status, we observed an interaction, in which APOE genotype was related to steeper decline in memory and language functioning in amyloid-β abnormal individuals only (β(SE)=-0.13(0.06); β(SE)=-0.22(0.07), respectively). CONCLUSION Our results suggest that APOE genotype is related to steeper decline in memory and language functioning in individuals with abnormal amyloid-β only. Furthermore, independent of amyloid-β status other genetic risk variants contribute to memory decline in initially cognitively unimpaired older individuals.
Collapse
Affiliation(s)
- Jori Tomassen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Amsterdam UMC location VUmc, 1007 MB, Amsterdam, PO Box 7057, The Netherlands.
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Elles Konijnenberg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Stephen F Carter
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Lyduine E Collij
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
104
|
Karaman BK, Mormino EC, Sabuncu MR. Machine learning based multi-modal prediction of future decline toward Alzheimer's disease: An empirical study. PLoS One 2022; 17:e0277322. [PMID: 36383528 PMCID: PMC9668188 DOI: 10.1371/journal.pone.0277322] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that progresses over decades. Early detection of individuals at high risk of future progression toward AD is likely to be of critical significance for the successful treatment and/or prevention of this devastating disease. In this paper, we present an empirical study to characterize how predictable an individual subjects' future AD trajectory is, several years in advance, based on rich multi-modal data, and using modern deep learning methods. Crucially, the machine learning strategy we propose can handle different future time horizons and can be trained with heterogeneous data that exhibit missingness and non-uniform follow-up visit times. Our experiments demonstrate that our strategy yields predictions that are more accurate than a model trained on a single time horizon (e.g. 3 years), which is common practice in prior literature. We also provide a comparison between linear and nonlinear models, verifying the well-established insight that the latter can offer a boost in performance. Our results also confirm that predicting future decline for cognitively normal (CN) individuals is more challenging than for individuals with mild cognitive impairment (MCI). Intriguingly, however, we discover that prediction accuracy decreases with increasing time horizon for CN subjects, but the trend is in the opposite direction for MCI subjects. Additionally, we quantify the contribution of different data types in prediction, which yields novel insights into the utility of different biomarkers. We find that molecular biomarkers are not as helpful for CN individuals as they are for MCI individuals, whereas magnetic resonance imaging biomarkers (hippocampus volume, specifically) offer a significant boost in prediction accuracy for CN individuals. Finally, we show how our model's prediction reveals the evolution of individual-level progression risk over a five-year time horizon. Our code is available at https://github.com/batuhankmkaraman/mlbasedad.
Collapse
Affiliation(s)
- Batuhan K. Karaman
- School of Electrical and Computer Engineering, Cornell University and Cornell Tech, New York, NY, United States of America
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States of America
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States of America
| | - Mert R. Sabuncu
- School of Electrical and Computer Engineering, Cornell University and Cornell Tech, New York, NY, United States of America
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States of America
| | | |
Collapse
|
105
|
Amyloidogenesis and Neurotrophic Dysfunction in Alzheimer’s Disease: Do They have a Common Regulating Pathway? Cells 2022; 11:cells11203201. [PMID: 36291068 PMCID: PMC9600014 DOI: 10.3390/cells11203201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
The amyloid cascade hypothesis has predominately been used to describe the pathogenesis of Alzheimer’s disease (AD) for decades, as Aβ oligomers are thought to be the prime cause of AD. Meanwhile, the neurotrophic factor hypothesis has also been proposed for decades. Accumulating evidence states that the amyloidogenic process and neurotrophic dysfunction are mutually influenced and may coincidently cause the onset and progress of AD. Meanwhile, there are intracellular regulators participating both in the amyloidogenic process and neurotrophic pathways, which might be the common original causes of amyloidogenesis and neurotrophic dysfunction. In this review, the current understanding regarding the role of neurotrophic dysfunction and the amyloidogenic process in AD pathology is briefly summarized. The mutual influence of these two pathogenesis pathways and their potential common causal pathway are further discussed. Therapeutic strategies targeting the common pathways to simultaneously prevent amyloidogenesis and neurotrophic dysfunction might be anticipated for the disease-modifying treatment of AD.
Collapse
|
106
|
Lin L, Ni L, Wang X, Sheng C. Longitudinal cognitive change and duration of Alzheimer's disease stages in relation to cognitive reserve. Neuroscience 2022; 504:47-55. [PMID: 36181989 DOI: 10.1016/j.neuroscience.2022.09.017] [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: 06/09/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 10/31/2022]
Abstract
The relationship of cognitive reserve and measures of reserve with longitudinal cognitive change and the duration of preclinical, prodromal, and mild Alzheimer disease (AD) dementia remains to be fully characterized. In our study, 660 β-amyloid-positive participants staged with preclinical AD, prodromal AD, and dementia due to AD from the Alzheimer's Disease Neuroimaging Initiative were selected. Cognitive reserve and brain reserve were defined by conventional proxies or the residual method at baseline. We evaluated the utility of these reserves in predicting longitudinal cognitive change by mixed effects models and used a multi-state model to estimate stage duration stratified by reserve groups. Corrected for age, sex, and APOE-ε4 status, reserve was associated with cognitive decline, and the effects changed depending on the specific measures of reserve and the stage of AD. Reserves defined by the residual method were stronger predictors of cognitive change than those defined by conventional proxies. The estimated time from preclinical to mild AD dementia varied from 15-24 years based on the different reserve groups, and we observed a linear trend for the longest duration in individuals with high cognitive reserve/high brain reserve, followed by those with high cognitive reserve/low brain reserve, low cognitive reserve/high brain reserve, and low cognitive reserve/low brain reserve. This study showed a reduced risk of cognitive decline for individuals with higher level of reserve regardless of methods for measuring reserve. Interindividual differences in reserve may be important for clinical practice and trial design.
Collapse
Affiliation(s)
- Li Lin
- Department of Radiology, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong, China.
| | - Lianghui Ni
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiaoni Wang
- Department of Neurology, Sir Run Run Shaw Hospital, Affiliated with School of Medicine, Zhejiang University, Hangzhou, China
| | - Can Sheng
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
107
|
Tomassen J, den Braber A, van der Landen SM, Konijnenberg E, Teunissen CE, Vermunt L, de Geus EJC, Boomsma DI, Scheltens P, Tijms BM, Visser PJ. Abnormal cerebrospinal fluid levels of amyloid and tau are associated with cognitive decline over time in cognitively normal older adults: A monozygotic twin study. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12346. [PMID: 36185992 PMCID: PMC9489168 DOI: 10.1002/trc2.12346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022]
Abstract
Introduction The contribution of genetic and environmental factors to the relation between cerebrospinal fluid (CSF) biomarkers and cognitive decline in preclinical Alzheimer's disease remains unclear. We studied this in initially cognitively normal monozygotic twins. Methods We included 122 cognitively normal monozygotic twins (51 pairs) with a follow-up of 4.3 ± 0.4 years. We first tested associations of baseline CSF Aβ1-42/1-40 ratio, total tau (t-tau), and 181-phosphorylated-tau (p-tau) status with subsequent cognitive decline using linear mixed models, and then performed twin specific analyses. Results Baseline abnormal amyloid-β and tau CSF markers predicted steeper decline on memory (p ≤ .003) and language (p ≤ 0.04). Amyloid-β and p-tau markers in one twin predicted decline in memory in the co-twin and tau markers in one twin predicted decline in language in the co-twin (r range -0.26,0.39; p's ≤ .02). Discussion These results suggest that memory and language decline are early features of AD that are in part determined by the same genetic factors that influence amyloid-β and tau regulation.
Collapse
Affiliation(s)
- Jori Tomassen
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
| | - Anouk den Braber
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Sophie M. van der Landen
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
| | - Elles Konijnenberg
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
| | - Charlotte E. Teunissen
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
- Neurochemistry Laboratory, Department of Clinical ChemistryVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Lisa Vermunt
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
- Neurochemistry Laboratory, Department of Clinical ChemistryVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eco J. C. de Geus
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Dorret I. Boomsma
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Philip Scheltens
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
| | - Betty M. Tijms
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center AmsterdamNeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Alzheimer Center Limburg, School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Department of NeurobiologyDivision of NeurogeriatricsKarolinska InstitutetCare Sciences and SocietyStockholmSweden
| |
Collapse
|
108
|
Park CJ, Seo Y, Choe YS, Jang H, Lee H, Kim JP. Predicting conversion of brain β-amyloid positivity in amyloid-negative individuals. Alzheimers Res Ther 2022; 14:129. [PMID: 36096822 PMCID: PMC9465850 DOI: 10.1186/s13195-022-01067-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022]
Abstract
Background Cortical deposition of β-amyloid (Aβ) plaque is one of the main hallmarks of Alzheimer’s disease (AD). While Aβ positivity has been the main concern so far, predicting whether Aβ (−) individuals will convert to Aβ (+) has become crucial in clinical and research aspects. In this study, we aimed to develop a classifier that predicts the conversion from Aβ (−) to Aβ (+) using artificial intelligence. Methods Data were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort regarding patients who were initially Aβ (−). We developed an artificial neural network-based classifier with baseline age, gender, APOE ε4 genotype, and global and regional standardized uptake value ratios (SUVRs) from positron emission tomography. Ten times repeated 10-fold cross-validation was performed for model measurement, and the feature importance was assessed. To validate the prediction model, we recruited subjects at the Samsung Medical Center (SMC). Results A total of 229 participants (53 converters) from the ADNI dataset and a total of 40 subjects (10 converters) from the SMC dataset were included. The average area under the receiver operating characteristic values of three developed models are as follows: Model 1 (age, gender, APOE ε4) of 0.674, Model 2 (age, gender, APOE ε4, global SUVR) of 0.814, and Model 3 (age, gender, APOE ε4, global and regional SUVR) of 0.841. External validation result showed an AUROC of 0.900. Conclusion We developed prediction models regarding Aβ positivity conversion. With the growing recognition of the need for earlier intervention in AD, the results of this study are expected to contribute to the screening of early treatment candidates. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01067-8.
Collapse
|
109
|
Cody KA, Koscik RL, Erickson CM, Berman SE, Jonaitis EM, Williams VJ, Mueller KD, Christian BT, Chin NA, Clark LR, Betthauser TJ, Johnson SC. Associations of the Lifestyle for Brain Health index with longitudinal cognition and brain amyloid beta in clinically unimpaired older adults: Findings from the Wisconsin Registry for Alzheimer's Prevention. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12351. [PMID: 36110432 PMCID: PMC9464997 DOI: 10.1002/dad2.12351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 01/25/2023]
Abstract
Introduction Modifiable health and lifestyle factors increase risk of dementia, but whether modifiable factors, when measured in late-midlife, impact the emergence or progression of Alzheimer's disease (AD) pathophysiologic or cognitive changes remains unresolved. Methods In initially cognitively unimpaired, late middle-aged participants (N = 1215; baseline age, M [standard deviation] = 59.3 [6.7] years) from the Wisconsin Registry for Alzheimer's Prevention (WRAP), we investigated the influence of the Lifestyle for Brain Health (LIBRA) index, a lifestyle-based dementia risk score, on AD-related cognitive trajectories and amyloid beta (Aβ) plaque accumulation. Results Overall, lower baseline LIBRA, denoting healthier lifestyle and lower dementia risk, was related to better overall cognitive performance, but did not moderate apolipoprotein E ε4 or Aβ-related longitudinal cognitive trajectories. LIBRA was not significantly associated with Aβ accumulation or estimated age of Aβ onset. Discussion In WRAP, late-midlife LIBRA scores were related to overall cognitive performance, but not AD-related cognitive decline or Aβ accumulation in the preclinical timeframe. Highlights The Lifestyle for Brain Health (LIBRA) index was associated with cognitive performance in late-midlife.LIBRA did not moderate apolipoprotein E ε4 or amyloid-related cognitive decline.LIBRA was not associated with the onset or accumulation of amyloid plaques.
Collapse
Affiliation(s)
- Karly A. Cody
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Rebecca L. Koscik
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Claire M. Erickson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sara E. Berman
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Erin M. Jonaitis
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Victoria J. Williams
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Kimberly D. Mueller
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Communication Sciences & DisordersUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Bradley T. Christian
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Waisman Laboratory for Brain Imaging and BehaviorUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Nathanial A. Chin
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Lindsay R. Clark
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton Veterans HospitalMadisonWisconsinUSA
| | - Tobey J. Betthauser
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton Veterans HospitalMadisonWisconsinUSA
| |
Collapse
|
110
|
Initial Data and a Clinical Diagnosis Transition for the Aiginition Longitudinal Biomarker Investigation of Neurodegeneration (ALBION) Study. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091179. [PMID: 36143858 PMCID: PMC9501022 DOI: 10.3390/medicina58091179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/26/2022]
Abstract
Background and Objectives: This article presents data from the ongoing Aiginition Longitudinal Biomarker Investigation of Neurodegeneration study (ALBION) regarding baseline clinical characterizations and CSF biomarker profiles, as well as preliminary longitudinal data on clinical progression. Materials and Methods: As of March 2022, 138 participants who either were cognitively normal (CN, n = 99) or had a diagnosis of mild cognitive impairment (MCI, n = 39) had been recruited at the specialist cognitive disorders outpatient clinic at Aiginition Hospital. Clinical characteristics at baseline were provided. These patients were followed annually to determine progression from CN to MCI or even dementia. CSF biomarker data (amyloid β1-42, phosphorylated tau at threonine 181, and total tau) collected using automated Elecsys® assays (Roche Diagnostics) were available for 74 patients. These patients were further sorted based on the AT(N) classification model, as determined by CSF Aβ42 (A), CSF pTau (T), and CSF tTau (N). Results: Of the 49 CN patients with CSF biomarker data, 21 (43%) were classified as exhibiting “Alzheimer’s pathologic change” (A+Τ− (Ν)−) and 6 (12%) as having “Alzheimer’s disease” (A+T−(N)+, A+T+(N)−, or A+T+(N)+). Of the 25 MCI patients, 8 (32%) displayed “Alzheimer’s pathologic change”, and 6 (24%) had “Alzheimer’s disease”. A total of 66 individuals had a mean follow-up of 2.1 years (SD = 0.9, min = 0.8, max = 3.9), and 15 of those individuals (22%) showed a clinical progression (defined as a worsening clinical classification, i.e., from CN to MCI or dementia or from MCI to dementia). Overall, participants with the “AD continuum” AT(N) biomarker profile (i.e., A+T−(N)−, A+T−(N)+, A+T+(N)−, and A+T+(N)+) were more likely to clinically progress (p = 0.04). Conclusions: A CSF “AD continuum” AT(N) biomarker profile is associated with an increased risk of future clinical decline in CN or MCI subjects.
Collapse
|
111
|
Torres-Espín A, Ferguson AR. Harmonization-Information Trade-Offs for Sharing Individual Participant Data in Biomedicine. HARVARD DATA SCIENCE REVIEW 2022; 4:10.1162/99608f92.a9717b34. [PMID: 36420049 PMCID: PMC9681014 DOI: 10.1162/99608f92.a9717b34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024] Open
Abstract
Biomedical practice is evidence-based. Peer-reviewed papers are the primary medium to present evidence and data-supported results to drive clinical practice. However, it could be argued that scientific literature does not contain data, but rather narratives about and summaries of data. Meta-analyses of published literature may produce biased conclusions due to the lack of transparency in data collection, publication bias, and inaccessibility to the data underlying a publication ('dark data'). Co-analysis of pooled data at the level of individual research participants can offer higher levels of evidence, but this requires that researchers share raw individual participant data (IPD). FAIR (findable, accessible, interoperable, and reusable) data governance principles aim to guide data lifecycle management by providing a framework for actionable data sharing. Here we discuss the implications of FAIR for data harmonization, an essential step for pooling data for IPD analysis. We describe the harmonization-information trade-off, which states that the level of granularity in harmonizing data determines the amount of information lost. Finally, we discuss a framework for managing the trade-off and the levels of harmonization. In the coming era of funder mandates for data sharing, research communities that effectively manage data harmonization will be empowered to harness big data and advanced analytics such as machine learning and artificial intelligence tools, leading to stunning new discoveries that augment our understanding of diseases and their treatments. By elevating scientific data to the status of a first-class citizen of the scientific enterprise, there is strong potential for biomedicine to transition from a narrative publication product orientation to a modern data-driven enterprise where data itself is viewed as a primary work product of biomedical research.
Collapse
Affiliation(s)
- Abel Torres-Espín
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, United States of America
| | - Adam R Ferguson
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, United States of America
- San Francisco Veterans Affairs Health Care System, San Francisco, California, United States of America
| |
Collapse
|
112
|
Johansson M, Stomrud E, Johansson PM, Svenningsson A, Palmqvist S, Janelidze S, van Westen D, Mattsson-Carlgren N, Hansson O. Development of Apathy, Anxiety, and Depression in Cognitively Unimpaired Older Adults: Effects of Alzheimer's Disease Pathology and Cognitive Decline. Biol Psychiatry 2022; 92:34-43. [PMID: 35346458 DOI: 10.1016/j.biopsych.2022.01.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/12/2022] [Accepted: 01/16/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND The impact of Alzheimer's disease (AD) pathology and cognitive deficits on longitudinal neuropsychiatric symptoms is unclear, especially in early disease stages. METHODS Cognitively unimpaired older adults (N = 356) enrolled in the prospective Swedish BioFINDER study were examined. Neuropsychiatric assessments encompassed the Apathy Evaluation Scale and the Hospital Anxiety and Depression Scale, performed biennially (together with tests of global cognition) for up to 8 years. Biomarkers were measured in cerebrospinal fluid or plasma at baseline. Magnetic resonance imaging quantified white matter lesions. We used linear mixed-effect models to test associations between baseline AD biomarkers (for amyloid-β [Aβ], tau, and neurodegeneration) and white matter lesions with longitudinal neuropsychiatric symptoms (apathy, anxiety, and depressive symptoms). We also tested associations between changes in cognition and changes in neuropsychiatric symptoms. Finally, we tested if change in cognition mediated the effects of different brain pathologies on neuropsychiatric symptoms. RESULTS Aβ pathology at baseline was associated with increasing levels of apathy (β = -0.284, p = .005) and anxiety (β = -0.060, p = .011) longitudinally. More rapid decline of cognition over time was related to increasing levels of apathy. The effects of baseline Aβ pathology on longitudinal apathy were partly mediated by changes in cognitive performance (proportion mediated 23%). CONCLUSIONS Aβ pathology may drive the development of both apathy and anxiety in very early stages of AD, largely independent of cognitive change. The effect of Aβ on apathy is only partially conveyed by worse cognition. Together, these findings highlight certain neuropsychiatric symptoms as early manifestations of AD.
Collapse
Affiliation(s)
- Maurits Johansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Division of Clinical Sciences Helsingborg, Department of Clinical Sciences Lund, Lund University, Helsingborg, Sweden; Clinical Department of Psychiatry, Helsingborg Hospital, Helsingborg, Sweden.
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Per Mårten Johansson
- Division of Clinical Sciences Helsingborg, Department of Clinical Sciences Lund, Lund University, Helsingborg, Sweden; Department of Internal Medicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Anna Svenningsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Image and Function, Skåne University Hospital Lund, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Neurology, Skåne University Hospital Lund, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
113
|
Lozupone M, Berardino G, Mollica A, Sardone R, Dibello V, Zupo R, Lampignano L, Castellana F, Bortone I, Stallone R, Daniele A, Altamura M, Bellomo A, Solfrizzi V, Panza F. ALZT-OP1: An experimental combination regimen for the treatment of Alzheimer's Disease. Expert Opin Investig Drugs 2022; 31:759-771. [PMID: 35758153 DOI: 10.1080/13543784.2022.2095261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION For Alzheimer's disease (AD) treatment, US FDA granted accelerated approval for aducanumab due to its amyloid-β (Aβ)-lowering effects, notwithstanding the reported poor correlation between amyloid plaque reduction and clinical change for this drug. The diversification of drug targets appears to be the future of the AD field and from this perspective, drugs modulating microglia dysfunction and combination treatment regimens offer some promise. AREAS COVERED The aim of the present article was to provide a comprehensive review of ALZT-OP1 (cromolyn sodium plus ibuprofen), an experimental combination treatment regimen for AD, discussing their mechanisms of action targeting Aβ and neuroinflammation, examining the role of microglia in AD and offering our own insights on the role of present and alternative approaches directed toward neuroinflammation. EXPERT OPINION Enrolling high-risk participants with elevated brain amyloid could help to slow cognitive decline in secondary prevention trials during AD preclinical stages. Long-term follow-up indicated that non-steroidal anti-inflammatory drugs use begun when the brain was still normal may benefit these patients, suggesting that the timing of therapy could be crucial. However, previous clinical failures and the present incomplete understanding of the Aβ pathophysiological role in AD put this novel experimental combination regimen at substantial risk of failure.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Berardino
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Anita Mollica
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Rodolfo Sardone
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Vittorio Dibello
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Roberta Zupo
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Luisa Lampignano
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Fabio Castellana
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Ilaria Bortone
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Roberta Stallone
- Neuroscience and Education, Human Resources Excellence in Research, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy.,Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Panza
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| |
Collapse
|
114
|
Morrison C, Dadar M, Villeneuve S, Collins DL. White matter lesions may be an early marker for age-related cognitive decline. Neuroimage Clin 2022; 35:103096. [PMID: 35764028 PMCID: PMC9241138 DOI: 10.1016/j.nicl.2022.103096] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/16/2022] [Accepted: 06/19/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Research suggests that cerebral small vessel disease (CSVD), amyloid, and pTau contribute to age-related cognitive decline. It remains unknown how these factors relate to one another and how they jointly contribute to cognitive decline in normal aging. This project examines the association between these factors and their relationship to cognitive decline in cognitively unimpaired older adults without subjective cognitive decline. METHODS A total of 230 subjects with cerebrospinal fluid (CSF) Aß42, CSF pTau181, white matter lesions (WMLs) used as a proxy of CSVD, and cognitive scores from the Alzheimer's Disease Neuroimaging Initiative were included. Associations between each factor and cognitive score were investigated using regression models. Furthermore, relationships between the three pathologies were also examined using regression models. RESULTS At baseline, there was an inverse association between WML load and Aß42 (t = -4.20, p <.001). There was no association between WML load and pTau (t = 0.32, p = 0.75), nor with Aß42 and pTau (t = 0.51, p =.61). Correcting for age, sex and education, baseline WML load was associated with baseline ADAS-13 scores (t = 2.59, p =.01) and lower follow-up executive functioning (t = -2.84, p =.005). Baseline Aß42 was associated with executive function at baseline (t = 3.58, p<.004) but not at follow-up (t = 1.05, p = 0.30), nor with ADAS-13 at baseline (t = -0.24, p = 0.81) or follow-up (t = 0.09, p = 0.93). Finally, baseline pTau was not associated with any cognitive measure at baseline or follow-up. CONCLUSION Both baseline Aß42 and WML load are associated with some baseline cognition scores, but only baseline WML load is associated with follow-up executive functioning. This finding suggests that WMLs may be one of the earliest clinical manifestations that contributes to future cognitive decline in cognitively healthy older adults. Given that healthy older adults with WMLs exhibit declines in cognitive functioning, they may be less resilient to future pathology increasing their risk for cognitive impairment due to dementia than those without WMLs.
Collapse
Affiliation(s)
- Cassandra Morrison
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, H3A 2B4 Montreal, Quebec, Canada.
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, H3A 1A1 Montreal, Quebec, Canada; Douglas Mental Health University Institute, Studies on Prevention of Alzheimer's Disease (StoP-AD) Centre, H4H 1R3 Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, H3A 2B4 Montreal, Quebec, Canada; Department of Psychiatry, McGill University, H3A 1A1 Montreal, Quebec, Canada; Douglas Mental Health University Institute, Studies on Prevention of Alzheimer's Disease (StoP-AD) Centre, H4H 1R3 Montreal, Quebec, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, H3A 2B4 Montreal, Quebec, Canada
| |
Collapse
|
115
|
Incremental diagnostic value of 18F-Fluetemetamol PET in differential diagnoses of Alzheimer's Disease-related neurodegenerative diseases from an unselected memory clinic cohort. Sci Rep 2022; 12:10385. [PMID: 35725910 PMCID: PMC9209498 DOI: 10.1038/s41598-022-14532-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022] Open
Abstract
To evaluate the incremental diagnostic value of 18F-Flutemetamol PET following MRI measurements on an unselected prospective cohort collected from a memory clinic. A total of 84 participants was included in this study. A stepwise study design was performed including initial analysis (based on clinical assessments), interim analysis (revision of initial analysis post-MRI) and final analysis (revision of interim analysis post-18F-Flutemetamol PET). At each time of evaluation, every participant was categorized into SCD, MCI or dementia syndromal group and further into AD-related, non-AD related or non-specific type etiological subgroup. Post 18F-Flutemetamol PET, the significant changes were seen in the syndromal MCI group (57%, p < 0.001) involving the following etiological subgroups: AD-related MCI (57%, p < 0.01) and non-specific MCI (100%, p < 0.0001); and syndromal dementia group (61%, p < 0.0001) consisting of non-specific dementia subgroup (100%, p < 0.0001). In the binary regression model, amyloid status significantly influenced the diagnostic results of interim analysis (p < 0.01). 18F-Flutemetamol PET can have incremental value following MRI measurements, particularly reflected in the change of diagnosis of individuals with unclear etiology and AD-related-suspected patients due to the role in complementing AD-related pathological information.
Collapse
|
116
|
Chapleau M, Iaccarino L, Soleimani-Meigooni D, Rabinovici GD. The Role of Amyloid PET in Imaging Neurodegenerative Disorders: A Review. J Nucl Med 2022; 63:13S-19S. [PMID: 35649652 DOI: 10.2967/jnumed.121.263195] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/22/2022] [Indexed: 12/17/2022] Open
Abstract
Imaging of amyloid deposition using PET has been available in research studies for 2 decades and has been approved for clinical use by the U.S. Food and Drug Administration, the European Medicines Agency, and other regulatory agencies around the world. Amyloid PET is a crucial tool for the diagnosis of Alzheimer disease, as it allows the noninvasive detection of amyloid plaques, a core neuropathologic feature that defines the disease. The clinical use of amyloid PET is expected to increase with recent accelerated approval in the United States of aducanumab, an antiamyloid monoclonal antibody, for the treatment of mild cognitive impairment and mild dementia due to Alzheimer disease. However, amyloid pathology can also be found in cognitively unimpaired older adults and in patients with other neurodegenerative disorders. The aim of this review is to provide an up-to-date overview of the application of amyloid PET in neurodegenerative diseases. We provide an in-depth analysis of the clinical, pathologic, and imaging correlates; a comparison with other available biomarkers; and a review of the application of amyloid PET in clinical trials and clinical utility studies.
Collapse
Affiliation(s)
- Marianne Chapleau
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California;
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - David Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California; and.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| |
Collapse
|
117
|
Sirkis DW, Bonham LW, Johnson TP, La Joie R, Yokoyama JS. Dissecting the clinical heterogeneity of early-onset Alzheimer's disease. Mol Psychiatry 2022; 27:2674-2688. [PMID: 35393555 PMCID: PMC9156414 DOI: 10.1038/s41380-022-01531-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) is a rare but particularly devastating form of AD. Though notable for its high degree of clinical heterogeneity, EOAD is defined by the same neuropathological hallmarks underlying the more common, late-onset form of AD. In this review, we describe the various clinical syndromes associated with EOAD, including the typical amnestic phenotype as well as atypical variants affecting visuospatial, language, executive, behavioral, and motor functions. We go on to highlight advances in fluid biomarker research and describe how molecular, structural, and functional neuroimaging can be used not only to improve EOAD diagnostic acumen but also enhance our understanding of fundamental pathobiological changes occurring years (and even decades) before the onset of symptoms. In addition, we discuss genetic variation underlying EOAD, including pathogenic variants responsible for the well-known mendelian forms of EOAD as well as variants that may increase risk for the much more common forms of EOAD that are either considered to be sporadic or lack a clear autosomal-dominant inheritance pattern. Intriguingly, specific pathogenic variants in PRNP and MAPT-genes which are more commonly associated with other neurodegenerative diseases-may provide unexpectedly important insights into the formation of AD tau pathology. Genetic analysis of the atypical clinical syndromes associated with EOAD will continue to be challenging given their rarity, but integration of fluid biomarker data, multimodal imaging, and various 'omics techniques and their application to the study of large, multicenter cohorts will enable future discoveries of fundamental mechanisms underlying the development of EOAD and its varied clinical presentations.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Taylor P Johnson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
118
|
Assunção SS, Sperling RA, Ritchie C, Kerwin DR, Aisen PS, Lansdall C, Atri A, Cummings J. Meaningful benefits: a framework to assess disease-modifying therapies in preclinical and early Alzheimer's disease. Alzheimers Res Ther 2022; 14:54. [PMID: 35440022 PMCID: PMC9017027 DOI: 10.1186/s13195-022-00984-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/05/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The need for preventive therapies that interrupt the progression of Alzheimer's disease (AD) before the onset of symptoms or when symptoms are emerging is urgent and has spurred the ongoing development of disease-modifying therapies (DMTs) in preclinical and early AD (mild cognitive impairment [MCI] to mild dementia). Assessing the meaningfulness of what are likely small initial treatment effects in these earlier stages of the AD patho-clinical disease continuum is a major challenge and warrants further consideration. BODY: To accommodate a shift towards earlier intervention in AD, we propose meaningful benefits as a new umbrella concept that encapsulates the spectrum of potentially desirable outcomes that may be demonstrated in clinical trials and other studies across the AD continuum, with an emphasis on preclinical AD and early AD (i.e., MCI due to AD and mild AD dementia). The meaningful benefits framework applies to data collection, assessment, and communication across three dimensions: (1) multidimensional clinical outcome assessments (COAs) including not only core disease outcomes related to cognition and function but also patient- and caregiver-reported outcomes, health and economic outcomes, and neuropsychiatric symptoms; (2) complementary analyses that help contextualize and expand the understanding of COA-based assessments, such as number-needed-to-treat or time-to-event analyses; and (3) assessment of both cumulative benefit and predictive benefit, where early changes on cognitive, functional, or biomarker assessments predict longer-term clinical benefit. CONCLUSION The concept of meaningful benefits emphasizes the importance of multidimensional reporting of clinical trial data while, conceptually, it advances our understanding of treatment effects in preclinical AD and mild cognitive impairment due to AD. We propose that such an approach will help bridge the gap between the emergence of DMTs and their clinical use, particularly now that a DMT is available for patients diagnosed with MCI due to AD and mild AD dementia.
Collapse
Affiliation(s)
- Sheila Seleri Assunção
- US Medical Affairs - Neuroscience, Genentech, A Member of the Roche Group, South San Francisco, CA, USA.
| | - Reisa A Sperling
- Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Scotland, UK
| | - Diana R Kerwin
- Kerwin Medical Center, Dallas, TX, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Paul S Aisen
- University of Southern California Alzheimer's Therapeutic Research Institute, San Diego, CA, USA
| | | | - Alireza Atri
- Banner Sun Health Research Institute, Sun City, AZ, USA
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV, USA
| |
Collapse
|
119
|
Tideman P, Stomrud E, Leuzy A, Mattsson-Carlgren N, Palmqvist S, Hansson O. Association of β-Amyloid Accumulation With Executive Function in Adults With Unimpaired Cognition. Neurology 2022; 98:e1525-e1533. [PMID: 35022305 PMCID: PMC9012270 DOI: 10.1212/wnl.0000000000013299] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 12/27/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The neuropathologic changes underlying Alzheimer disease (AD) start before overt cognitive symptoms arise, but it is not well-known how they relate to the first subtle cognitive changes. The objective for this study was to examine the independent associations of the AD hallmarks β-amyloid (Aβ), tau, and neurodegeneration with different cognitive domains in cognitively unimpaired (CU) individuals. METHODS In this cross-sectional study, CU participants from the prospective BioFINDER-2 study were included. All had CSF biomarkers (Aβ42 and phosphorylated tau [p-tau]181), MRI (cortical thickness of AD-susceptible regions), Aβ-PET (neocortical uptake), tau-PET (entorhinal uptake), and cognitive test data for memory, executive function, verbal function, and visuospatial function. Multivariable linear regression models were performed using either CSF Aβ42, p-tau181, and cortical thickness or Aβ-PET, tau-PET, and cortical thickness as predictors of cognitive function. The results were validated in an independent cohort (Alzheimer's Disease Neuroimaging Initiative [ADNI]). RESULTS A total of 316 CU participants were included from the BioFINDER-2 study. Abnormal Aβ status was independently associated with the executive measure, regardless of modality (CSF Aβ42, β = 0.128, p = 0.024; Aβ-PET, β = 0.124, p = 0.049), while tau was independently associated with memory (CSF p-tau181, β = 0.132, p = 0.018; tau-PET, β = 0.189, p = 0.002). Cortical thickness was independently associated with the executive measure and verbal fluency in both models (p = 0.005-0.018). To examine the relationships in the earliest stage of preclinical AD, only participants with normal biomarkers of tau and neurodegeneration were included (n = 217 CSF-based; n = 246 PET-based). Again, Aβ status was associated with executive function (CSF Aβ42, β = 0.189, p = 0.005; Aβ-PET, β = 0.146, p = 0.023), but not with other cognitive domains. The results were overall replicated in the ADNI cohort (n = 361). DISCUSSION These findings suggest that Aβ is independently associated with worse performance on an executive measure but not with memory performance, which instead is associated with tau pathology. This may have implications for early preclinical AD screening and outcome measures in AD trials targeting Aβ pathology.
Collapse
Affiliation(s)
- Pontus Tideman
- From the Clinical Memory Research Unit, Department of Clinical Sciences (P.T., E.S., A.L., N.M.-C., S.P., O.H.), and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; and Memory Clinic (P.T., E.S., S.P., O.H.) and Department of Neurology (N.M.-C.), Skåne University Hospital, Sweden
| | - Erik Stomrud
- From the Clinical Memory Research Unit, Department of Clinical Sciences (P.T., E.S., A.L., N.M.-C., S.P., O.H.), and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; and Memory Clinic (P.T., E.S., S.P., O.H.) and Department of Neurology (N.M.-C.), Skåne University Hospital, Sweden
| | - Antoine Leuzy
- From the Clinical Memory Research Unit, Department of Clinical Sciences (P.T., E.S., A.L., N.M.-C., S.P., O.H.), and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; and Memory Clinic (P.T., E.S., S.P., O.H.) and Department of Neurology (N.M.-C.), Skåne University Hospital, Sweden
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit, Department of Clinical Sciences (P.T., E.S., A.L., N.M.-C., S.P., O.H.), and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; and Memory Clinic (P.T., E.S., S.P., O.H.) and Department of Neurology (N.M.-C.), Skåne University Hospital, Sweden
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit, Department of Clinical Sciences (P.T., E.S., A.L., N.M.-C., S.P., O.H.), and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; and Memory Clinic (P.T., E.S., S.P., O.H.) and Department of Neurology (N.M.-C.), Skåne University Hospital, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit, Department of Clinical Sciences (P.T., E.S., A.L., N.M.-C., S.P., O.H.), and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; and Memory Clinic (P.T., E.S., S.P., O.H.) and Department of Neurology (N.M.-C.), Skåne University Hospital, Sweden.
| |
Collapse
|
120
|
Aisen PS, Jimenez-Maggiora GA, Rafii MS, Walter S, Raman R. Early-stage Alzheimer disease: getting trial-ready. Nat Rev Neurol 2022; 18:389-399. [PMID: 35379951 PMCID: PMC8978175 DOI: 10.1038/s41582-022-00645-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Slowing the progression of Alzheimer disease (AD) might be the greatest unmet medical need of our time. Although one AD therapeutic has received a controversial accelerated approval from the FDA, more effective and accessible therapies are urgently needed. Consensus is growing that for meaningful disease modification in AD, therapeutic intervention must be initiated at very early (preclinical or prodromal) stages of the disease. Although the methods for such early-stage clinical trials have been developed, identification and recruitment of the required asymptomatic or minimally symptomatic study participants takes many years and requires substantial funds. As an example, in the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s Disease Trial (the first phase III trial to be performed in preclinical AD), 3.5 years and more than 5,900 screens were required to recruit and randomize 1,169 participants. A new clinical trials infrastructure is required to increase the efficiency of recruitment and accelerate therapeutic progress. Collaborations in North America, Europe and Asia are now addressing this need by establishing trial-ready cohorts of individuals with preclinical and prodromal AD. These collaborations are employing innovative methods to engage the target population, assess risk of brain amyloid accumulation, select participants for biomarker studies and determine eligibility for trials. In the future, these programmes could provide effective tools for pursuing the primary prevention of AD. Here, we review the lessons learned from the AD trial-ready cohorts that have been established to date, with the aim of informing ongoing and future efforts towards efficient, cost-effective trial recruitment. Consensus is growing that intervention in the very early stages of Alzheimer disease is necessary for disease modification. Here, the authors discuss the challenges of recruiting asymptomatic or mildly symptomatic participants for clinical trials, focusing on ‘trial-ready’ cohorts as a potential solution. Trial-ready cohorts are an effective strategy for the identification of participants eligible for clinical trials in early-stage Alzheimer disease (AD). Building these cohorts requires considerable planning and technological infrastructure to facilitate recruitment, remote longitudinal assessment, data management and data storage. Trial-ready cohorts exist for genetically determined populations at risk of AD, such as those with familial AD and Down syndrome; the longitudinal data from these cohorts is improving our understanding of the disease progression in early stages, informing clinical trial design and accelerating recruitment to intervention studies. So far, the challenges experienced by trial-ready cohorts for early-stage AD have included difficulties recruiting an ethnically and racially representative cohort; and for online cohorts, difficulty retaining participants. The results of ongoing work will reveal the success of strategies to improve cohort diversity and retention, and the rates of referral to clinical trials.
Collapse
|
121
|
Li TR, Yang Q, Hu X, Han Y. Biomarkers and Tools for Predicting Alzheimer's Disease in the Preclinical Stage. Curr Neuropharmacol 2022; 20:713-737. [PMID: 34030620 PMCID: PMC9878962 DOI: 10.2174/1570159x19666210524153901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the only leading cause of death for which no disease-modifying therapy is currently available. Over the past decade, a string of disappointing clinical trial results has forced us to shift our focus to the preclinical stage of AD, which represents the most promising therapeutic window. However, the accurate diagnosis of preclinical AD requires the presence of brain β- amyloid deposition determined by cerebrospinal fluid or amyloid-positron emission tomography, significantly limiting routine screening and diagnosis in non-tertiary hospital settings. Thus, an easily accessible marker or tool with high sensitivity and specificity is highly needed. Recently, it has been discovered that individuals in the late stage of preclinical AD may not be truly "asymptomatic" in that they may have already developed subtle or subjective cognitive decline. In addition, advances in bloodderived biomarker studies have also allowed the detection of pathologic changes in preclinical AD. Exosomes, as cell-to-cell communication messengers, can reflect the functional changes of their source cell. Methodological advances have made it possible to extract brain-derived exosomes from peripheral blood, making exosomes an emerging biomarker carrier and liquid biopsy tool for preclinical AD. The eye and its associated structures have rich sensory-motor innervation. In this regard, studies have indicated that they may also provide reliable markers. Here, our report covers the current state of knowledge of neuropsychological and eye tests as screening tools for preclinical AD and assesses the value of blood and brain-derived exosomes as carriers of biomarkers in conjunction with the current diagnostic paradigm.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xiaochen Hu
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, 50924, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China;,Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China;,National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China;,School of Biomedical Engineering, Hainan University, Haikou, 570228, China;,Address correspondence to this author at the Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China; Tel: +86 13621011941; E-mail:
| |
Collapse
|
122
|
Marianetti M, Pinna S, Venuti A, Liguri G. Olive polyphenols and bioavailable glutathione: Promising results in patients diagnosed with mild Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12278. [PMID: 35310529 PMCID: PMC8918095 DOI: 10.1002/trc2.12278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 11/25/2022]
Abstract
Introduction Recent studies highlighted the role of olive polyphenols in disrupting the ordered structure of highly cytotoxic amyloid beta protofibrils and the efficacy of a derivatized form of glutathione to counteract neuronal oxidative stress affecting specific brain regions at early stages of Alzheimer's disease (AD) pathogenesis. We performed a randomized cross-over clinical trial to evaluate their potential benefits in mild AD. Methods Oleuropein and S-acetyl glutathione were administered as dietary supplement for 6 months to 18 patients diagnosed for probable mild AD according to International Working Group 2 criteria. Patients underwent an extensive cognitive and behavioral neuropsychological test battery at the beginning and end of the study to evaluate cognitive deterioration, memory, visuospatial abilities, attention, language, executive functions, and behavioral disorders. We compared patients receiving treatment to patients receiving no treatment. Results All the measured neurocognitive parameters stabilized or improved after the treatment in all patients. Discussion Dietary supplement with olive polyphenols and bioavailable glutathione could be useful for patients diagnosed with mild AD.
Collapse
Affiliation(s)
| | - Silvia Pinna
- Experimental Alzheimer CenterFatebenefratelli Roman ProvinceRomeItaly
| | - Angelo Venuti
- Experimental Alzheimer CenterFatebenefratelli Roman ProvinceRomeItaly
| | - Gianfranco Liguri
- Department of Biochemistry and Molecular BiologyUniversity of FlorenceFlorenceItaly
| |
Collapse
|
123
|
|
124
|
Kagerer SM, Schroeder C, van Bergen JMG, Schreiner SJ, Meyer R, Steininger SC, Vionnet L, Gietl AF, Treyer V, Buck A, Pruessmann KP, Hock C, Unschuld PG. Low Subicular Volume as an Indicator of Dementia-Risk Susceptibility in Old Age. Front Aging Neurosci 2022; 14:811146. [PMID: 35309894 PMCID: PMC8926841 DOI: 10.3389/fnagi.2022.811146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Hippocampal atrophy is an established Alzheimer’s Disease (AD) biomarker. Volume loss in specific subregions as measurable with ultra-high field magnetic resonance imaging (MRI) may reflect earliest pathological alterations. Methods Data from positron emission tomography (PET) for estimation of cortical amyloid β (Aβ) and high-resolution 7 Tesla T1 MRI for assessment of hippocampal subfield volumes were analyzed in 61 non-demented elderly individuals who were divided into risk-categories as defined by high levels of cortical Aβ and low performance in standardized episodic memory tasks. Results High cortical Aβ and low episodic memory interactively predicted subicular volume [F(3,57) = 5.90, p = 0.018]. The combination of high cortical Aβ and low episodic memory was associated with significantly lower subicular volumes, when compared to participants with high episodic memory (p = 0.004). Discussion Our results suggest that low subicular volume is linked to established indicators of AD risk, such as increased cortical Aβ and low episodic memory. Our data support subicular volume as a marker of dementia-risk susceptibility in old-aged non-demented persons.
Collapse
Affiliation(s)
- Sonja M. Kagerer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Clemens Schroeder
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | | | - Simon J. Schreiner
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Rafael Meyer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Stefanie C. Steininger
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Laetitia Vionnet
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Anton F. Gietl
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Klaas P. Pruessmann
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren, Switzerland
| | - Paul G. Unschuld
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
- Geriatric Psychiatry, Department of Psychiatry, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
- *Correspondence: Paul G. Unschuld,
| |
Collapse
|
125
|
Jack CR, Therneau TM, Lundt ES, Wiste HJ, Mielke MM, Knopman DS, Graff-Radford J, Lowe VJ, Vemuri P, Schwarz CG, Senjem ML, Gunter JL, Petersen RC. Long-term associations between amyloid positron emission tomography, sex, apolipoprotein E and incident dementia and mortality among individuals without dementia: hazard ratios and absolute risk. Brain Commun 2022; 4:fcac017. [PMID: 35310829 PMCID: PMC8924651 DOI: 10.1093/braincomms/fcac017] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/08/2021] [Accepted: 01/31/2022] [Indexed: 11/14/2022] Open
Abstract
Dementia and mortality rates rise inexorably with age and consequently interact. However, because of the major logistical difficulties in accounting for both outcomes in a defined population, very little work has examined how risk factors and biomarkers for incident dementia are influenced by competing mortality. The objective of this study was to examine long-term associations between amyloid PET, APOE ɛ4, sex, education and cardiovascular/metabolic conditions, and hazard and absolute risk of dementia and mortality in individuals without dementia at enrolment. Participants were enrolled in the Mayo Clinic Study of Aging, a population-based study of cognitive ageing in Olmsted County, MN, USA. All were without dementia and were age 55-92 years at enrolment and were followed longitudinally. Predictor variables were amyloid PET, APOE ɛ4 status, sex, education, cardiovascular/metabolic conditions and age. The main outcomes were incident dementia and mortality. Multivariable, multi-state models were used to estimate mortality and incident dementia rates and absolute risk of dementia and mortality by predictor variable group. Of the 4984 participants in the study, 4336 (87%) were cognitively unimpaired and 648 (13%) had mild cognitive impairment at enrolment. The median age at enrolment was 75 years; 2463 (49%) were women. The median follow-up time was 9.4 years (7.5 years after PET). High versus normal amyloid (hazard ratio 2.11, 95% confidence interval 1.43-2.79), APOE ɛ4 (women: hazard ratio 2.24, 95% confidence interval 1.80-2.77; men: hazard ratio 1.37, 95% confidence interval 1.09-1.71), older age and two additional cardiovascular/metabolic conditions (hazard ratio 1.37, 95% confidence interval 1.22-1.53) were associated with the increased hazard of dementia (all P < 0.001). Among APOE ɛ4 carriers with elevated amyloid, remaining lifetime risk of dementia at age 65 years was greater in women [74% (95% confidence interval 65-84%) high and 58% (95% confidence interval 52-65%) moderate amyloid], than men [62% (95% confidence interval 52-73%) high and 44% (95% confidence interval 35-53%) moderate amyloid]. Overall, the hazard and absolute risk of dementia varied considerably by predictor group. The absolute risk of dementia associated with predictors characteristic of Alzheimer's disease was greater in women than men while at the same time the combination of APOE ɛ4 non-carrier with normal amyloid was more protective in women than men. This set of findings may be attributed in part to different biological effects and in part to lower mortality rates in women.
Collapse
Affiliation(s)
| | - Terry M. Therneau
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Emily S. Lundt
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - 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
| | | | | | - Val J. Lowe
- Department of Nuclear Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | |
Collapse
|
126
|
Affiliation(s)
- Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
127
|
Luo W, Wen H, Ge S, Tang C, Liu X, Lu L. Development of a Sex-Specific Risk Scoring System for the Prediction of Cognitively Normal People to Patients With Mild Cognitive Impairment (SRSS-CNMCI). Front Aging Neurosci 2022; 13:774804. [PMID: 35145390 PMCID: PMC8823413 DOI: 10.3389/fnagi.2021.774804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE We aimed to develop a sex-specific risk scoring system, abbreviated as SRSS-CNMCI, for the prediction of the conversion of cognitively normal (CN) people into patients with Mild Cognitive Impairment (MCI) to provide a reliable tool for the prevention of MCI. METHODS CN at baseline participants 61-90 years of age were selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database with at least one follow-up. Multivariable Cox proportional hazards models were used to identify the major risk factors associated with the conversion from CN to MCI and to develop the SRSS-CNMCI. Receiver operating characteristic (ROC) curve analysis was used to determine risk cutoff points corresponding to an optimal prediction. The results were externally validated, including evaluation of the discrimination and calibration in the Harvard Aging Brain Study (HABS) database. RESULTS A total of 471 participants, including 240 female (51%) and 231 male participants (49%) aged from 61 to 90 years, were included in the study cohort. The final multivariable models and the SRSS-CNMCI included age, APOE e4, mini mental state examination (MMSE) and clinical dementia rating (CDR). The C-statistics of the SRSS-CNMCI were 0.902 in the female subgroup and 0.911 in the male subgroup. The cutoff point of high and low risks was 33% in the female subgroup, indicating that more than 33% female participants were considered to have a high risk, and more than 9% participants were considered to have a high risk in the male subgroup. The SRSS-CNMCI performed well in the external cohort: the C-statistics were 0.950 in the female subgroup and 0.965 in the male subgroup. CONCLUSION The SRSS-CNMCI performs well in various cohorts and provides an accurate prediction and a generalization.
Collapse
Affiliation(s)
- Wen Luo
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
- Evidence-Based Medicine and Data Science Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Wen
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuqi Ge
- Evidence-Based Medicine and Data Science Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunzhi Tang
- Evidence-Based Medicine and Data Science Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiufeng Liu
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liming Lu
- Evidence-Based Medicine and Data Science Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
128
|
Ferguson LA, Leal SL. Interactions of Emotion and Memory in the Aging Brain: Neural and Psychological Correlates. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-021-00245-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
129
|
Gao Q, Daunt P, Gibson AM, Pither RJ. Utility of Polygenic Risk Scoring to Predict Cognitive Impairment as Measured by Preclinical Alzheimer Cognitive Composite Score. JAR LIFE 2022; 11:1-8. [PMID: 36923235 PMCID: PMC10002888 DOI: 10.14283/jarlife.2022.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 03/18/2023]
Abstract
Background The utility of Polygenic Risk Scores (PRS) is gaining increasing attention for generating an individual genetic risk profile to predict subsequent likelihood of future onset of Alzheimer's disease (AD), especially those carry two copies of the APOE E3 allele, currently considered at neutral risk in all populations studied. Objectives To access the performance of PRS in predicting individuals whilst pre-symptomatic or with mild cognitive impairment who are at greatest risk of progression of cognitive impairment due to Alzheimer's Disease from the Alzheimer's Disease Neuroimaging Initiative (ADNI) as measured by the Preclinical Alzheimer Cognitive Composite (PACC) score profile. Design: A longitudinal analysis of data from the ADNI study conducted across over 50 sites in the US and Canada. Setting Multi-centre genetics study. Participants 594 subjects either APOE E3 homozygotes or APOE E3/E4 heterozygotes who upon entry to the study were diagnosed as cognitively normal or with mild cognitive impairment. Measurements Use of genotyping and/or whole genome sequencing data to calculate polygenic risk scores and assess its ability to predict subsequent cognitive decline as measured by PACC over 5 years. Results: Assessing both cognitively normal and mild cognitive impaired subjects using a PRS threshold of greater than 0.6, the high genetic risk participant group declined more than the low risk group over 5 years as measured by PACC score (PACC score reduced by time). Conclusions Our findings have shown that polygenic risk score provides a promising tool to identify those with higher risk to decline over 5 years regardless of their APOE alleles according to modified PACC profile, especially its ability to identify APOE3/E3 cognitively normal individuals who are at most risk for early cognitive decline. This genotype accounts for approximately 60% of the general population and 35% of the AD population but currently would not be considered at higher risk without access to expensive or invasive biomarker testing.
Collapse
Affiliation(s)
- Q Gao
- Cytox Limited, Manchester, UK
| | - P Daunt
- Cytox Limited, Manchester, UK
| | | | | | | |
Collapse
|
130
|
Amyloid and tau PET-positive cognitively unimpaired individuals are at high risk for future cognitive decline. Nat Med 2022; 28:2381-2387. [PMID: 36357681 PMCID: PMC9671808 DOI: 10.1038/s41591-022-02049-x] [Citation(s) in RCA: 199] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/21/2022] [Indexed: 11/12/2022]
Abstract
A major unanswered question in the dementia field is whether cognitively unimpaired individuals who harbor both Alzheimer's disease neuropathological hallmarks (that is, amyloid-β plaques and tau neurofibrillary tangles) can preserve their cognition over time or are destined to decline. In this large multicenter amyloid and tau positron emission tomography (PET) study (n = 1,325), we examined the risk for future progression to mild cognitive impairment and the rate of cognitive decline over time among cognitively unimpaired individuals who were amyloid PET-positive (A+) and tau PET-positive (T+) in the medial temporal lobe (A+TMTL+) and/or in the temporal neocortex (A+TNEO-T+) and compared them with A+T- and A-T- groups. Cox proportional-hazards models showed a substantially increased risk for progression to mild cognitive impairment in the A+TNEO-T+ (hazard ratio (HR) = 19.2, 95% confidence interval (CI) = 10.9-33.7), A+TMTL+ (HR = 14.6, 95% CI = 8.1-26.4) and A+T- (HR = 2.4, 95% CI = 1.4-4.3) groups versus the A-T- (reference) group. Both A+TMTL+ (HR = 6.0, 95% CI = 3.4-10.6) and A+TNEO-T+ (HR = 7.9, 95% CI = 4.7-13.5) groups also showed faster clinical progression to mild cognitive impairment than the A+T- group. Linear mixed-effect models indicated that the A+TNEO-T+ (β = -0.056 ± 0.005, T = -11.55, P < 0.001), A+TMTL+ (β = -0.024 ± 0.005, T = -4.72, P < 0.001) and A+T- (β = -0.008 ± 0.002, T = -3.46, P < 0.001) groups showed significantly faster longitudinal global cognitive decline compared to the A-T- (reference) group (all P < 0.001). Both A+TNEO-T+ (P < 0.001) and A+TMTL+ (P = 0.002) groups also progressed faster than the A+T- group. In summary, evidence of advanced Alzheimer's disease pathological changes provided by a combination of abnormal amyloid and tau PET examinations is strongly associated with short-term (that is, 3-5 years) cognitive decline in cognitively unimpaired individuals and is therefore of high clinical relevance.
Collapse
|
131
|
Ma LZ, Hu H, Wang ZT, Ou YN, Dong Q, Tan L, Yu JT. P-tau and neurodegeneration mediate the effect of β-amyloid on cognition in non-demented elders. Alzheimers Res Ther 2021; 13:200. [PMID: 34911582 PMCID: PMC8675473 DOI: 10.1186/s13195-021-00943-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/05/2021] [Indexed: 04/12/2023]
Abstract
BACKGROUND There are many pathological changes in the brains of Alzheimer's disease (AD) patients. For many years, the mainstream view on the pathogenesis of AD believes that β-amyloid (Aβ) usually acts independently in addition to triggering functions. However, the evidence now accumulating indicates another case that these pathological types have synergies. The objective of this study was to investigate whether effects of Aβ pathology on cognition were mediated by AD pathologies, including tau-related pathology (p-tau), neurodegeneration (t-tau, MRI measurements), axonal injury (NFL), synaptic dysfunction (neurogranin), and neuroinflammation (sTREM2, YKL-40). METHODS Three hundred seventy normal controls (CN) and 623 MCI patients from the ADNI (Alzheimer's Disease Neuroimaging Initiative) database were recruited in this research. Linear mixed-effects models were used to evaluate the associations of baseline Aβ with cognitive decline and biomarkers of several pathophysiological pathways. Causal mediation analyses with 10,000 bootstrapped iterations were conducted to explore the mediation effects of AD pathologies on cognition. RESULTS Tau-related pathology, neurodegeneration, neuroinflammation are correlated with the concentration of Aβ, even in CN participants. The results show that age, gender, and APOE ε4 carrier status have a moderating influence on some of these relationships. There is a stronger association of Aβ with biomarkers and cognitive changes in the elderly and females. In CN group, Aβ pathology is directly related to poor cognition and has no mediating effect (p < 0.05). In mild cognitive impairment, tau-related pathology (26.15% of total effect) and neurodegeneration (14.8% to 47.0% of total effect) mediate the impact of Aβ on cognition. CONCLUSIONS In conclusion, early Aβ accumulation has an independent effect on cognitive decline in CN and a tau, neurodegeneration-dependent effect in the subsequent cognitive decline in MCI patients.
Collapse
Affiliation(s)
- Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
132
|
Decreased Resting-State Functional Complexity in Elderly with Subjective Cognitive Decline. ENTROPY 2021; 23:e23121591. [PMID: 34945897 PMCID: PMC8700613 DOI: 10.3390/e23121591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022]
Abstract
Individuals with subjective cognitive decline (SCD) are at high risk of developing preclinical or clinical state of Alzheimer’s disease (AD). Resting state functional magnetic resonance imaging, which can indirectly reflect neuron activities by measuring the blood-oxygen-level-dependent (BOLD) signals, is promising in the early detection of SCD. This study aimed to explore whether the nonlinear complexity of BOLD signals can describe the subtle differences between SCD and normal aging, and uncover the underlying neuropsychological implications of these differences. In particular, we introduce amplitude-aware permutation entropy (AAPE) as the novel measure of brain entropy to characterize the complexity in BOLD signals in each brain region of the Brainnetome atlas. Our results demonstrate that AAPE can reflect the subtle differences between both groups, and the SCD group presented significantly decreased complexities in subregions of the superior temporal gyrus, the inferior parietal lobule, the postcentral gyrus, and the insular gyrus. Moreover, the results further reveal that lower complexity in SCD may correspond to poorer cognitive performance or even subtle cognitive impairment. Our findings demonstrated the effectiveness and sensitiveness of the novel brain entropy measured by AAPE, which may serve as the potential neuroimaging marker for exploring the subtle changes in SCD.
Collapse
|
133
|
Chan MY, Han L, Carreno CA, Zhang Z, Rodriguez RM, LaRose M, Hassenstab J, Wig GS. Long-term prognosis and educational determinants of brain network decline in older adult individuals. NATURE AGING 2021; 1:1053-1067. [PMID: 35382259 PMCID: PMC8979545 DOI: 10.1038/s43587-021-00125-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022]
Abstract
Older adults with lower education are at greater risk for dementia. It is unclear which brain changes lead to these outcomes. Longitudinal imaging-based measures of brain structure and function were examined in adult individuals (baseline age, 45-86 years; two to five visits per participant over 1-9 years). College degree completion differentiates individual-based and neighborhood-based measures of socioeconomic status and disadvantage. Older adults (~65 years and over) without a college degree exhibit a pattern of declining large-scale functional brain network organization (resting-state system segregation) that is less evident in their college-educated peers. Declining brain system segregation predicts impending changes in dementia severity, measured up to 10 years past the last scan date. The prognostic value of brain network change is independent of Alzheimer's disease (AD)-related genetic risk (APOE status), the presence of AD-associated pathology (cerebrospinal fluid phosphorylated tau, cortical amyloid) and cortical thinning. These results demonstrate that the trajectory of an individual's brain network organization varies in relation to their educational attainment and, more broadly, is a unique indicator of individual brain health during older age.
Collapse
Affiliation(s)
- Micaela Y. Chan
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Liang Han
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Claudia A. Carreno
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Ziwei Zhang
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Rebekah M. Rodriguez
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Megan LaRose
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gagan S. Wig
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
134
|
Deng Y, Zhu H, Xiao L, Liu C, Liu YL, Gao W. Identification of the function and mechanism of m6A reader IGF2BP2 in Alzheimer's disease. Aging (Albany NY) 2021; 13:24086-24100. [PMID: 34705667 PMCID: PMC8610118 DOI: 10.18632/aging.203652] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/03/2021] [Indexed: 01/06/2023]
Abstract
Alzheimer’s disease, the most common form of dementia in the elderly, is a kind of neurodegenerative disease. However, its pathogenesis and diagnosis remain unclear. M6A is related to nervous system development and neurodegenerative diseases. Here in this study, using multiple RNA-seq datasets of Alzheimer’s brain tissues, along with bioinformatic analysis, we innovatively found that m6A reader protein IGF2BP2 was abnormally highly expressed in Alzheimer’s patients. After compared between Alzheimer’s and normal brain samples, and between IGF2BP2- high and IGF2BP2- low subgroups of Alzheimer’s patients, we took the shared differentially expressed genes as the relevant gene sets of IGF2PB2 affecting Alzheimer’s disease occurrence for subsequent analysis. Then, weight gene correlation analysis was conducted and 17 functional modules were identified. The module that most positively correlated with Alzheimer’s disease and IGF2PB2-high subgroups were mainly participated in ECM receptor interaction, focal adhesion, cytokine-cytokine receptor interaction, and TGF-beta signaling pathway. Afterwards, a hub gene-based model including 20 genes was constructed by LASSO regression and validated by ROC curve for Alzheimer diagnosis. Finally, we preliminarily elucidated that IGF2BP2 could bind with mRNAs in a m6A-dependent manner. This study first elucidates the pathogenic role of IGF2BP2 in Alzheimer’s disease. IGF2BP2 and its relevant m6A modifications are potential to be new diagnostic and therapeutic targets for Alzheimer’s patients.
Collapse
Affiliation(s)
- Yanyao Deng
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan Province, China
| | - Hongwei Zhu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Le Xiao
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan Province, China
| | - Chao Liu
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan Province, China
| | - Ya-Lin Liu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China.,Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Wenzhe Gao
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
135
|
Li QS, Vasanthakumar A, Davis JW, Idler KB, Nho K, Waring JF, Saykin AJ. Association of peripheral blood DNA methylation level with Alzheimer's disease progression. Clin Epigenetics 2021; 13:191. [PMID: 34654479 PMCID: PMC8518178 DOI: 10.1186/s13148-021-01179-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background Identifying biomarkers associated with Alzheimer’s disease (AD) progression may enable patient enrichment and improve clinical trial designs. Epigenome-wide association studies have revealed correlations between DNA methylation at cytosine-phosphate-guanine (CpG) sites and AD pathology and diagnosis. Here, we report relationships between peripheral blood DNA methylation profiles measured using Infinium® MethylationEPIC BeadChip and AD progression in participants from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort. Results The rate of cognitive decline from initial DNA sampling visit to subsequent visits was estimated by the slopes of the modified Preclinical Alzheimer Cognitive Composite (mPACC; mPACCdigit and mPACCtrailsB) and Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) plots using robust linear regression in cognitively normal (CN) participants and patients with mild cognitive impairment (MCI), respectively. In addition, diagnosis conversion status was assessed using a dichotomized endpoint. Two CpG sites were significantly associated with the slope of mPACC in CN participants (P < 5.79 × 10−8 [Bonferroni correction threshold]); cg00386386 was associated with the slope of mPACCdigit, and cg09422696 annotated to RP11-661A12.5 was associated with the slope of CDR-SB. No significant CpG sites associated with diagnosis conversion status were identified. Genes involved in cognition and learning were enriched. A total of 19, 13, and 5 differentially methylated regions (DMRs) associated with the slopes of mPACCtrailsB, mPACCdigit, and CDR-SB, respectively, were identified by both comb-p and DMRcate algorithms; these included DMRs annotated to HOXA4. Furthermore, 5 and 19 DMRs were associated with conversion status in CN and MCI participants, respectively. The most significant DMR was annotated to the AD-associated gene PM20D1 (chr1: 205,818,956 to 205,820,014 [13 probes], Sidak-corrected P = 7.74 × 10−24), which was associated with both the slope of CDR-SB and the MCI conversion status. Conclusion Candidate CpG sites and regions in peripheral blood were identified as associated with the rate of cognitive decline in participants in the ADNI cohort. While we did not identify a single CpG site with sufficient clinical utility to be used by itself due to the observed effect size, a biosignature composed of DNA methylation changes may have utility as a prognostic biomarker for AD progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01179-2.
Collapse
Affiliation(s)
- Qingqin S Li
- Neuroscience, Janssen Research and Development, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ, 08560, USA.
| | | | - Justin W Davis
- Genomics Research Center, AbbVie, North Chicago, IL, USA
| | | | - Kwangsik Nho
- Indiana Alzheimer's Disease Research Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
136
|
Musiek ES, Gomez-Isla T, Holtzman DM. Aducanumab for Alzheimer disease: the amyloid hypothesis moves from bench to bedside. J Clin Invest 2021; 131:e154889. [PMID: 34651585 PMCID: PMC8516468 DOI: 10.1172/jci154889] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Erik S. Musiek
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Teresa Gomez-Isla
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Massachusetts Alzheimer’s Disease Research Center, Boston, Massachusetts, USA
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
137
|
Relationships between frontal metabolites and Alzheimer's disease biomarkers in cognitively normal older adults. Neurobiol Aging 2021; 109:22-30. [PMID: 34638000 DOI: 10.1016/j.neurobiolaging.2021.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 12/18/2022]
Abstract
Elevated expression of β-amyloid (Aβ1-42) and tau are considered risk-factors for Alzheimer's disease in healthy older adults. We investigated the effect of aging and cerebrospinal fluid levels of Aβ1-42 and tau on 1) frontal metabolites measured with proton magnetic resonance spectroscopy (MRS) and 2) cognition in cognitively normal older adults (n = 144; age range 50-85). Levels of frontal gamma aminobutyric acid (GABA+) and myo-inositol relative to creatine (mI/tCr) were predicted by age. Levels of GABA+ predicted cognitive performance better than mI/tCr. Additionally, we found that frontal levels of n-acetylaspartate relative to creatine (tNAA/tCr) were predicted by levels of t-tau. In cognitively normal older adults, levels of frontal GABA+ and mI/tCr are predicted by aging, with levels of GABA+ decreasing with age and the opposite for mI/tCr. These results suggest that age- and biomarker-related changes in brain metabolites are not only located in the posterior cortex as suggested by previous studies and further demonstrate that MRS is a viable tool in the study of aging and biomarkers associated with pathological aging and Alzheimer's disease.
Collapse
|
138
|
Genetic effects on longitudinal cognitive decline during the early stages of Alzheimer's disease. Sci Rep 2021; 11:19853. [PMID: 34615922 PMCID: PMC8494841 DOI: 10.1038/s41598-021-99310-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 09/22/2021] [Indexed: 11/08/2022] Open
Abstract
Cognitive decline in early-stage Alzheimer's disease (AD) may depend on genetic variability. In the Swedish BioFINDER study, we used polygenic scores (PGS) (for AD, intelligence, and educational attainment) to predict longitudinal cognitive change (measured by mini-mental state examination (MMSE) [primary outcome] and other cognitive tests) over a mean of 4.2 years. We included 260 β-amyloid (Aβ) negative cognitively unimpaired (CU) individuals, 121 Aβ-positive CU (preclinical AD), 50 Aβ-negative mild cognitive impairment (MCI) patients, and 127 Aβ-positive MCI patients (prodromal AD). Statistical significance was determined at Bonferroni corrected p value < 0.05. The PGS for intelligence (beta = 0.1, p = 2.9e-02) was protective against decline in MMSE in CU and MCI participants regardless of Aβ status. The polygenic risk score for AD (beta = - 0.12, p = 9.4e-03) was correlated with the rate of change in MMSE and was partially mediated by Aβ-pathology (mediation effect 20%). There was no effect of education PGS on cognitive measures. Genetic variants associated with intelligence mitigate cognitive decline independent of Aβ-pathology, while effects of genetic variants associated with AD are partly mediated by Aβ-pathology.
Collapse
|
139
|
Apolipoprotein E ɛ4-related effects on cognition are limited to the Alzheimer's disease spectrum. GeroScience 2021; 44:195-209. [PMID: 34591236 PMCID: PMC8811053 DOI: 10.1007/s11357-021-00450-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/23/2021] [Indexed: 11/29/2022] Open
Abstract
Whether the deleterious effects of APOE4 are restricted to the Alzheimer’s disease (AD) spectrum or cause cognitive impairment irrespectively of the development of AD is still a matter of debate, and the focus of this study. Our analyses included APOE4 genotype, neuropsychological variables, amyloid-βeta (Aβ) and Tau markers, FDG-PET values, and hippocampal volumetry data derived from the healthy controls sample of the ADNI database. We formed 4 groups of equal size (n = 30) based on APOE4 carriage and amyloid-PET status. Baseline and follow-up (i.e., 48 months post-baseline) results indicated that Aβ-positivity was the most important factor to explain poorer cognitive performance, while APOE4 only exerted a significant effect in Aβ-positive subjects. Additionally, multiple regression analyses evidenced that, within the Aβ-positive sample, hippocampal volumetry explained most of the variability in cognitive performance for APOE4 carriers. These findings represent a strong support for the so-called preclinical/prodromal hypothesis, which states that the reported differences in cognitive performance between healthy carriers and non-carriers are mainly due to the APOE4’s capability to increase the risk of AD. Moreover, our results reinforce the notion that a synergistic interaction of Aβ and APOE4 elicits a neurodegenerative process in the hippocampus that might be the main cause of impaired cognitive performance.
Collapse
|
140
|
Sexton C, Snyder H, Beher D, Boxer AL, Brannelly P, Brion JP, Buée L, Cacace AM, Chételat G, Citron M, DeVos SL, Diaz K, Feldman HH, Frost B, Goate AM, Gold M, Hyman B, Johnson K, Karch CM, Kerwin DR, Koroshetz WJ, Litvan I, Morris HR, Mummery CJ, Mutamba J, Patterson MC, Quiroz YT, Rabinovici GD, Rommel A, Shulman MB, Toledo-Sherman LM, Weninger S, Wildsmith KR, Worley SL, Carrillo MC. Current directions in tau research: Highlights from Tau 2020. Alzheimers Dement 2021; 18:988-1007. [PMID: 34581500 DOI: 10.1002/alz.12452] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 11/07/2022]
Abstract
Studies supporting a strong association between tau deposition and neuronal loss, neurodegeneration, and cognitive decline have heightened the allure of tau and tau-related mechanisms as therapeutic targets. In February 2020, leading tau experts from around the world convened for the first-ever Tau2020 Global Conference in Washington, DC, co-organized and cosponsored by the Rainwater Charitable Foundation, the Alzheimer's Association, and CurePSP. Representing academia, industry, government, and the philanthropic sector, presenters and attendees discussed recent advances and current directions in tau research. The meeting provided a unique opportunity to move tau research forward by fostering global partnerships among academia, industry, and other stakeholders and by providing support for new drug discovery programs, groundbreaking research, and emerging tau researchers. The meeting also provided an opportunity for experts to present critical research-advancing tools and insights that are now rapidly accelerating the pace of tau research.
Collapse
Affiliation(s)
| | | | | | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Pat Brannelly
- Alzheimer's Disease Data Initiative, Kirkland, WI, USA
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Luc Buée
- Univ Lille, Inserm, CHU-Lille, Lille Neuroscience and Cognition, Place de Verdun, Lille, France
| | | | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, France
| | - Martin Citron
- Neuroscience TA, Braine l'Alleud, UCB Biopharma, Brussels, Belgium
| | - Sarah L DeVos
- Translational Sciences, Denali Therapeutics, San Francisco, California, USA
| | | | - Howard H Feldman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Bess Frost
- Sam & Ann Barshop Institute for Longevity and Aging Studies, Glenn Biggs Institute for Alzheimer's & Neurodegenerative Disorders, Department of Cell Systems & Anatomy, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Neuroscience, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Gold
- AbbVie, Neurosciences Development, North Chicago, Illinois, USA
| | - Bradley Hyman
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Johnson
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Diana R Kerwin
- Kerwin Medical Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Walter J Koroshetz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, San Diego, California, USA
| | - Huw R Morris
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | - Catherine J Mummery
- Dementia Research Centre, National Hospital for Neurology and Neurosurgery, University College London, London, UK
| | | | - Marc C Patterson
- Departments of Neurology, Pediatrics and Medical Genetics, Mayo Clinic, Rochester, Minnesota, USA
| | - Yakeel T Quiroz
- Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Departments of Neurology, Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Amy Rommel
- Tau Consortium, Rainwater Charitable Foundation, Fort Worth, Texas, USA
| | - Melanie B Shulman
- Neurodegeneration Development Unit, Biogen, Boston, Massachusetts, USA
| | | | | | - Kristin R Wildsmith
- Department of Biomarker Development, Genentech, South San Francisco, California, USA
| | - Susan L Worley
- Independent science writer, Bryn Mawr, Pennsylvania, USA
| | | |
Collapse
|
141
|
Jacobs HI, Becker JA, Kwong K, Engels-Domínguez N, Prokopiou PC, Papp KV, Properzi M, Hampton OL, Uquillas FD, Sanchez JS, Rentz DM, Fakhri GE, Normandin MD, Price JC, Bennett DA, Sperling RA, Johnson KA. In vivo and neuropathology data support locus coeruleus integrity as indicator of Alzheimer's disease pathology and cognitive decline. Sci Transl Med 2021; 13:eabj2511. [PMID: 34550726 PMCID: PMC8641759 DOI: 10.1126/scitranslmed.abj2511] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several autopsy studies recognize the locus coeruleus (LC) as the initial site of hyperphosphorylated TAU aggregation, and as the number of LC neurons harboring TAU increases, TAU pathology emerges throughout the cortex. By conjointly using dedicated MRI measures of LC integrity and TAU and amyloid PET imaging, we aimed to address the question whether in vivo LC measures relate to initial cortical patterns of Alzheimer’s disease (AD) fibrillar proteinopathies or cognitive dysfunction in 174 cognitively unimpaired and impaired older individuals with longitudinal cognitive measures. To guide our interpretations, we verified these associations in autopsy data from 1524 Religious Orders Study and Rush Memory and Aging Project and 2145 National Alzheimer’s Coordinating Center cases providing three different LC measures (pigmentation, tangle density, and neuronal density), Braak staging, β-amyloid, and longitudinal cognitive measures. Lower LC integrity was associated with elevated TAU deposition in the entorhinal cortex among unimpaired individuals consistent with postmortem correlations between LC tangle density and successive Braak staging. LC pigmentation ratings correlated with LC neuronal density but not with LC tangle density and were particularly worse at advanced Braak stages. In the context of elevated β-amyloid, lower LC integrity and greater cortical tangle density were associated with greater TAU burden beyond the medial temporal lobe and retrospective memory decline. These findings support neuropathologic data in which early LC TAU accumulation relates to disease progression and identify LC integrity as a promising indicator of initial AD-related processes and subtle changes in cognitive trajectories of preclinical AD.
Collapse
Affiliation(s)
- Heidi I.L. Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
- Harvard Medical School; Boston, MA, 02115, USA
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University; 6200MD Maastricht, The Netherlands
| | - John A. Becker
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
- Harvard Medical School; Boston, MA, 02115, USA
| | - Kenneth Kwong
- Harvard Medical School; Boston, MA, 02115, USA
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02129, USA
| | - Nina Engels-Domínguez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University; 6200MD Maastricht, The Netherlands
| | - Prokopis C. Prokopiou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
- Harvard Medical School; Boston, MA, 02115, USA
| | - Kathryn V. Papp
- Harvard Medical School; Boston, MA, 02115, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital; Boston, MA,02115, USA
| | - Michael Properzi
- Harvard Medical School; Boston, MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital; Boston, MA, 02114, USA
| | - Olivia L. Hampton
- Department of Neurology, Massachusetts General Hospital; Boston, MA, 02114, USA
| | | | - Justin S. Sanchez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
| | - Dorene M. Rentz
- Harvard Medical School; Boston, MA, 02115, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital; Boston, MA,02115, USA
- Department of Neurology, Massachusetts General Hospital; Boston, MA, 02114, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
- Harvard Medical School; Boston, MA, 02115, USA
| | - Marc D. Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
- Harvard Medical School; Boston, MA, 02115, USA
| | - Julie C. Price
- Harvard Medical School; Boston, MA, 02115, USA
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02129, USA
| | - David A. Bennett
- Department of Neurological Sciences, Rush Alzheimer’s Disease Center, Rush University Medical Center; Chicago, Illinois, 60612, USA
| | - Reisa A. Sperling
- Harvard Medical School; Boston, MA, 02115, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital; Boston, MA,02115, USA
- Department of Neurology, Massachusetts General Hospital; Boston, MA, 02114, USA
| | - Keith A. Johnson
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital; Boston, MA, 02114, USA
- Harvard Medical School; Boston, MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital; Boston, MA, 02114, USA
| |
Collapse
|
142
|
Zhou R, Chen LL, Yang H, Li L, Liu J, Chen L, Hong WJ, Wang CG, Ma JJ, Huang J, Zhou XF, Liu D, Zhou HD. Effect of High Cholesterol Regulation of LRP1 and RAGE on Aβ Transport Across the Blood-Brain Barrier in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:428-442. [PMID: 34488598 DOI: 10.2174/1567205018666210906092940] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 04/01/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND High cholesterol aggravates the risk development of Alzheimer's disease (AD). AD is closely related to the transport impairment of Amyloid-β (Aβ) in the blood-brain barrier. It is unclear whether high cholesterol affects the risk of cognitive impairment in AD by affecting Aβ transport. The purpose of the study is to investigate whether high cholesterol regulates Aβ transport through low-density Lipoprotein Receptor-Related Protein 1 (LRP1) and Receptor for Advanced Glycation End products (RAGE) in the risk development of AD. METHODS We established high cholesterol AD mice model. The learning and memory functions were evaluated by Morris Water Maze (MWM). Cerebral microvascular endothelial cells were isolated, cultured, and observed. The expression levels of LRP1 and RAGE of endothelial cells and their effect on Aβ transport in vivo were observed. The expression level of LRP1 and RAGE was detected in cultured microvessels after using Wnt inhibitor DKK-1 and β-catenin inhibitor XAV-939. RESULTS Hypercholesterolemia exacerbated spatial learning and memory impairment. Hypercholesterolemia increased serum Aβ40 level, while serum Aβ42 level did not change significantly. Hypercholesterolemia decreased LRP1 expression and increased RAGE expression in cerebral microvascular endothelial cells. Hypercholesterolemia increased brain apoptosis in AD mice. In in vitro experiment, high cholesterol decreased LRP1 expression and increased RAGE expression, increased Aβ40 expression in cerebral microvascular endothelial cells. High cholesterol regulated the expressions of LRP1 and RAGE and transcriptional activity of LRP1 and RAGE promoters by the Wnt/β-catenin signaling pathway. CONCLUSION High cholesterol decreased LRP1 expression and increased RAGE expression in cerebral microvascular endothelial cells, which led to Aβ transport disorder in the blood-brain barrier. Increased Aβ deposition in the brain aggravated apoptosis in the brain, resulting to cognitive impairment of AD mice.
Collapse
Affiliation(s)
- Rui Zhou
- Department of Orthopedics, The Orthopedic Surgery Center of Chinese PLA, Southwest Hospital, Army Medical University, Chongqing 400042, China
| | - Li-Li Chen
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Hai Yang
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ling Li
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Juan Liu
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Le Chen
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Wen-Juan Hong
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Cong-Guo Wang
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Jing-Jing Ma
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Jie Huang
- Postgraduate School, Bengbu Medical College, Anhui 233004, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences and Sansom Institute, University of South Australia, Adelaide, SA, Australia
| | - Dong Liu
- Laboratory of Field Surgery Institute, Army Medical University, Chongqing 400042, China
| | - Hua-Dong Zhou
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
143
|
Walden LM, Hu S, Madabhushi A, Prescott JW. Amyloid Deposition Is Greater in Cerebral Gyri than in Cerebral Sulci with Worsening Clinical Diagnosis Across the Alzheimer's Disease Spectrum. J Alzheimers Dis 2021; 83:423-433. [PMID: 34334397 DOI: 10.3233/jad-210308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Histopathologic studies have demonstrated differential amyloid-β (Aβ) burden between cortical sulci and gyri in Alzheimer's disease (AD), with sulci having a greater Aβ burden. OBJECTIVE To characterize Aβ deposition in the sulci and gyri of the cerebral cortex in vivo among subjects with normal cognition (NC), mild cognitive impairment (MCI), and AD, and to evaluate if these differences could improve discrimination between diagnostic groups. METHODS T1-weighted 3T MR and florbetapir (amyloid) positron emission tomography (PET) data were obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI). T1 images were segmented and the cortex was separated into sulci/gyri based on pial surface curvature measurements. T1 images were registered to PET images and regional standardized uptake value ratios (SUVr) were calculated. A linear mixed effects model was used to analyze the relationship between clinical variables and amyloid PET SUVr measurements in the sulci/gyri. Receiver operating characteristic (ROC) analysis was performed to define amyloid positivity. Logistic models were used to evaluate predictive performance of clinical diagnosis using amyloid PET SUVr measurements in sulci/gyri. RESULTS 719 subjects were included: 272 NC, 315 MCI, and 132 AD. Gyral and sulcal Aβ increased with worsening cognition, however there was a greater increase in gyral Aβ. Females had a greater gyral and sulcal Aβ burden. Focusing on sulcal and gyral Aβ did not improve predictive power for diagnostic groups. CONCLUSION While there were significant differences in Aβ deposition in cerebral sulci and gyri across the AD spectrum, these differences did not translate into improved prediction of diagnosis. Females were found to have greater gyral and sulcal Aβ burden.
Collapse
Affiliation(s)
- Lucas M Walden
- MetroHealth, Department of Radiology, Cleveland, OH, USA
| | - Song Hu
- MetroHealth, Department of Radiology, Cleveland, OH, USA
| | - Anant Madabhushi
- Case Western Reserve University, Department of Biomedical Engineering, Center for Computational Imaging & Personalized Diagnostics, Cleveland, OH, USA.,Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Jeffrey W Prescott
- MetroHealth, Department of Radiology, Cleveland, OH, USA.,Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | | |
Collapse
|
144
|
Sevinc G, Rusche J, Wong B, Datta T, Kaufman R, Gutz SE, Schneider M, Todorova N, Gaser C, Thomalla G, Rentz D, Dickerson BD, Lazar SW. Mindfulness Training Improves Cognition and Strengthens Intrinsic Connectivity Between the Hippocampus and Posteromedial Cortex in Healthy Older Adults. Front Aging Neurosci 2021; 13:702796. [PMID: 34512305 PMCID: PMC8430251 DOI: 10.3389/fnagi.2021.702796] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023] Open
Abstract
Maintaining optimal cognitive functioning throughout the lifespan is a public health priority. Evaluation of cognitive outcomes following interventions to promote and preserve brain structure and function in older adults, and associated neural mechanisms, are therefore of critical importance. In this randomized controlled trial, we examined the behavioral and neural outcomes following mindfulness training (n = 72), compared to a cognitive fitness program (n = 74) in healthy, cognitively normal, older adults (65-80 years old). To assess cognitive functioning, we used the Preclinical Alzheimer Cognitive Composite (PACC), which combines measures of episodic memory, executive function, and global cognition. We hypothesized that mindfulness training would enhance cognition, increase intrinsic functional connectivity measured with magnetic resonance imaging (MRI) between the hippocampus and posteromedial cortex, as well as promote increased gray matter volume within those regions. Following the 8-week intervention, the mindfulness training group showed improved performance on the PACC, while the control group did not. Furthermore, following mindfulness training, greater improvement on the PACC was associated with a larger increase in intrinsic connectivity within the default mode network, particularly between the right hippocampus and posteromedial cortex and between the left hippocampus and lateral parietal cortex. The cognitive fitness training group did not show such effects. These findings demonstrate that mindfulness training improves cognitive performance in cognitively intact older individuals and strengthens connectivity within the default mode network, which is particularly vulnerable to aging affects. Clinical Trial Registration: [https://clinicaltrials.gov/ct2/show/NCT02628548], identifier [NCT02628548].
Collapse
Affiliation(s)
- Gunes Sevinc
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Johann Rusche
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Kopf- und Neurozentrum, Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bonnie Wong
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Tanya Datta
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Robert Kaufman
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sarah E. Gutz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA, United States
| | - Marissa Schneider
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Nevyana Todorova
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Behavioral Neuroscience, College of Science, Northeastern University, Boston, MA, United States
| | - Christian Gaser
- Department of Psychiatry and Neurology, Jena University Hospital, Jena, Germany
| | - Götz Thomalla
- Kopf- und Neurozentrum, Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorene Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Bradford D. Dickerson
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sara W. Lazar
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
145
|
Dougherty RJ, Ramachandran J, Liu F, An Y, Wanigatunga AA, Tian Q, Bilgel M, Simonsick EM, Ferrucci L, Resnick SM, Schrack JA. Association of walking energetics with amyloid beta status: Findings from the Baltimore Longitudinal Study of Aging. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12228. [PMID: 34458552 PMCID: PMC8377776 DOI: 10.1002/dad2.12228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Higher energetic costs for mobility predict gait speed decline. Slow gait is linked to cognitive decline and Alzheimer's disease (AD). Whether the energetic cost of walking is linked to AD pathology is unknown. We investigated the cross-sectional association between the energetic cost of walking, gait speed, and amyloid beta (Aβ) status (+/-) in older adults. METHODS One hundred forty-nine cognitively normal adults (56% women, mean age 77.5 ± 8.4 years) completed customary-paced walking assessments with indirect calorimetry and 11C-Pittsburgh compound B positron emission tomography. Logistic regression models examined associations adjusted for demographics, body composition, comorbid conditions, and apolipoprotein E ε4. RESULTS Each 0.01 mL/kg/m greater energy cost was associated with 18% higher odds of being Aβ+ (odds ratio [OR] = 1.18; 95% confidence interval [CI]: 1.04 to 1.34; P = .011). These findings were not observed when investigating gait speed (OR = 0.99; 95% CI: 0.97 to 1.01; P = .321). DISCUSSION High energetic cost of walking is linked to AD pathology and may be a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Ryan J. Dougherty
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Janani Ramachandran
- Departments of Epidemiology and BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Fangyu Liu
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Yang An
- Intramural Research Program, National Institute on AgingBaltimoreMarylandUSA
| | - Amal A. Wanigatunga
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Center on Aging and HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Qu Tian
- Intramural Research Program, National Institute on AgingBaltimoreMarylandUSA
| | - Murat Bilgel
- Intramural Research Program, National Institute on AgingBaltimoreMarylandUSA
| | | | - Luigi Ferrucci
- Intramural Research Program, National Institute on AgingBaltimoreMarylandUSA
| | - Susan M. Resnick
- Intramural Research Program, National Institute on AgingBaltimoreMarylandUSA
| | - Jennifer A. Schrack
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Intramural Research Program, National Institute on AgingBaltimoreMarylandUSA
- Center on Aging and HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
146
|
Longitudinal analysis of APOE-ε4 genotype with the logical memory delayed recall score in Alzheimer’s disease. J Genet 2021. [DOI: 10.1007/s12041-021-01309-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
147
|
Espay AJ, Sturchio A, Schneider LS, Ezzat K. Soluble Amyloid-β Consumption in Alzheimer's Disease. J Alzheimers Dis 2021; 82:1403-1415. [PMID: 34151810 DOI: 10.3233/jad-210415] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain proteins function in their soluble, native conformation and cease to function when transformed into insoluble aggregates, also known as amyloids. Biophysically, the soluble-to-insoluble phase transformation represents a process of polymerization, similar to crystallization, dependent on such extrinsic factors as concentration, pH, and a nucleation surface. The resulting cross-β conformation of the insoluble amyloid is markedly stable, making it an unlikely source of toxicity. The spread of brain amyloidosis can be fully explained by mechanisms of spontaneous or catalyzed polymerization and phase transformation instead of active replication, which is an enzyme- and energy-requiring process dependent on a specific nucleic acid code for the transfer of biological information with high fidelity. Early neuronal toxicity in Alzheimer's disease may therefore be mediated to a greater extent by a reduction in the pool of soluble, normal-functioning protein than its accumulation in the polymerized state. This alternative loss-of-function hypothesis of pathogenicity can be examined by assessing the clinical and neuroimaging effects of administering non-aggregating peptide analogs to replace soluble amyloid-β levels above the threshold below which neuronal toxicity may occur. Correcting the depletion of soluble amyloid-β, however, would only exemplify 'rescue medicine.' Precision medicine will necessitate identifying the pathogenic factors catalyzing the protein aggregation in each affected individual. Only then can we stratify patients for etiology-specific treatments and launch precision medicine for Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andrea Sturchio
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Lon S Schneider
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kariem Ezzat
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
148
|
CSF Proteomic Alzheimer's Disease-Predictive Subtypes in Cognitively Intact Amyloid Negative Individuals. Proteomes 2021; 9:proteomes9030036. [PMID: 34449748 PMCID: PMC8396164 DOI: 10.3390/proteomes9030036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/10/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022] Open
Abstract
We recently discovered three distinct pathophysiological subtypes in Alzheimer’s disease (AD) using cerebrospinal fluid (CSF) proteomics: one with neuronal hyperplasticity, a second with innate immune system activation, and a third subtype with blood–brain barrier dysfunction. It remains unclear whether AD proteomic subtype profiles are a consequence of amyloid aggregation, or might exist upstream from aggregated amyloid. We studied this question in 127 older individuals with intact cognition and normal AD biomarkers in two independent cohorts (EMIF-AD MBD and ADNI). We clustered 705 proteins measured in CSF that were previously related to AD. We identified in these cognitively intact individuals without AD pathology three subtypes: two subtypes were seen in both cohorts (n = 49 with neuronal hyperplasticity and n = 44 with blood–brain barrier dysfunction), and one only in ADNI (n = 12 with innate immune activation). The proteins specific for these subtypes strongly overlapped with AD subtype protein profiles (overlap coefficients 92%–71%). Longitudinal p181-tau and amyloid β 1–42 (Aβ42) CSF analysis showed that in the hyperplasticity subtype p181-tau increased (β = 2.6 pg/mL per year, p = 0.01) and Aβ42 decreased over time (β = −4.4 pg/mL per year, p = 0.03), in the innate immune activation subtype p181-tau increased (β = 3.1 pg/mL per year, p = 0.01) while in the blood–brain barrier dysfunction subtype Aβ42 decreased (β = −3.7 pg/mL per year, p = 0.009). These findings suggest that AD proteomic subtypes might already manifest in cognitively normal individuals and may predispose for AD before amyloid has reached abnormal levels.
Collapse
|
149
|
Morgello S, Cortes EP, Gensler G, Meloni G, Jacobs MM, Murray J, Borukov V, Crary JF. HIV disease duration, but not active brain infection, predicts cortical amyloid beta deposition. AIDS 2021; 35:1403-1412. [PMID: 33813555 PMCID: PMC8243827 DOI: 10.1097/qad.0000000000002893] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Abnormal deposition of the antimicrobial peptide amyloid beta (Aβ) is a characteristic of Alzheimer's disease. The objective of this study was to elucidate risk factors for brain Aβ in a cohort enriched for HIV and other neurotropic pathogens. DESIGN Cross-sectional cohort study. METHODS We examined autopsy brains of 257 donors with a mean age of 52.8 years; 62% were men; and 194 were HIV+ and 63 HIV-. Hyperphosphorylated tau (p-tau) and Aβ were identified in frontal and temporal regions by immunohistochemistry. APOE genotyping was performed. Clinical and neuropathological predictors for Aβ were identified in univariate analyses, and then tested in multivariate regressions. RESULTS Cortical Aβ was identified in 32% of the sample, and active brain infection in 27%. Increased odds of Aβ were seen with increasing age and having an APOE ε4 allele; for the overall sample, HIV+ status was protective and brain infection was not a predictor. Within the HIV+ population, predictors for Aβ were duration of HIV disease and APOE alleles, but not age. When HIV disease duration and other HIV parameters were introduced into models for the entire sample, HIV disease duration was equivalent to age as a predictor of Aβ. CONCLUSION We hypothesize that dual aspects of immune suppression and stimulation in HIV, and beneficial survivor effects in older HIV+ individuals, account for HIV+ status decreasing, and HIV duration increasing, odds of Aβ. Importantly, with HIV, disease duration replaces age as an independent risk for Aβ, suggesting HIV-associated accelerated brain senescence.
Collapse
Affiliation(s)
- Susan Morgello
- Department of Neurology
- Department of Neuroscience
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Etty P Cortes
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York City, NY
| | | | | | | | | | - Valeriy Borukov
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York City, NY
| | - John F Crary
- Department of Neuroscience
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York City, NY
| |
Collapse
|
150
|
Ge X, Zhang D, Qiao Y, Zhang J, Xu J, Zheng Y. Association of Tau Pathology With Clinical Symptoms in the Subfields of Hippocampal Formation. Front Aging Neurosci 2021; 13:672077. [PMID: 34335226 PMCID: PMC8317580 DOI: 10.3389/fnagi.2021.672077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/20/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: To delineate the relationship between clinical symptoms and tauopathy of the hippocampal subfields under different amyloid statuses. Methods: One hundred and forty-three subjects were obtained from the ADNI project, including 87 individuals with normal cognition, 46 with mild cognitive impairment, and 10 with Alzheimer's disease (AD). All subjects underwent the tau PET, amyloid PET, T1W, and high-resolution T2W scans. Clinical symptoms were assessed by the Neuropsychiatric Inventory (NPI) total score and Alzheimer's Disease Assessment Scale cognition 13 (ADAS-cog-13) total score, comprising memory and executive function scores. The hippocampal subfields including Cornu Ammonis (CA1-3), subiculum (Sub), and dentate gyrus (DG), as well as the adjacent para-hippocampus (PHC) and entorhinal cortex (ERC), were segmented automatically using the Automatic Segmentation of Hippocampal Subfields (ASHS) software. The relationship between tauopathy/volume of the hippocampal subfields and assessment scores was calculated using partial correlation analysis under different amyloid status, by controlling age, gender, education, apolipoprotein E (APOE) allele ɛ4 carrier status, and, time interval between the acquisition time of tau PET and amyloid PET scans. Results: Compared with amyloid negative (A-) group, individuals from amyloid positive (A+) group are more impaired based on the Mini-mental State Examination (MMSE; p = 3.82e-05), memory (p = 6.30e-04), executive function (p = 0.0016), and ADAS-cog-13 scores (p = 5.11e-04). Significant decrease of volume (CA1, DG, and Sub) and increase of tau deposition (CA1, Sub, ERC, and PHC) of the hippocampal subfields of both hemispheres were observed for the A+ group compared to the A- group. Tauopathy of ERC is significantly associated with memory score for the A- group, and the associated regions spread into Sub and PHC for the A+ group. The relationship between the impairment of behavior or executive function and tauopathy of the hippocampal subfield was discovered within the A+ group. Leftward asymmetry was observed with the association between assessment scores and tauopathy of the hippocampal subfield, which is more prominent for the NPI score for the A+ group. Conclusion: The associations of tauopathy/volume of the hippocampal subfields with clinical symptoms provide additional insight into the understanding of local changes of the human HF during the AD continuum and can be used as a reference for future studies.
Collapse
Affiliation(s)
- Xinting Ge
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Dan Zhang
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yuchuan Qiao
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jiong Zhang
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Junhai Xu
- College of Intelligence and Computing, Tianjin Key Lab of Cognitive Computing and Application, Tianjin University, Tianjin, China
| | - Yuanjie Zheng
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
| |
Collapse
|